101
|
Gil-Gil T, Corona F, Martínez JL, Bernardini A. The Inactivation of Enzymes Belonging to the Central Carbon Metabolism Is a Novel Mechanism of Developing Antibiotic Resistance. mSystems 2020; 5:e00282-20. [PMID: 32487742 PMCID: PMC8534728 DOI: 10.1128/msystems.00282-20] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 05/14/2020] [Indexed: 11/20/2022] Open
Abstract
Fosfomycin is a bactericidal antibiotic, analogous to phosphoenolpyruvate, that exerts its activity by inhibiting the activity of MurA. This enzyme catalyzes the first step of peptidoglycan biosynthesis, the transfer of enolpyruvate from phosphoenolpyruvate to uridine-diphosphate-N-acetylglucosamine. Fosfomycin is increasingly being used, mainly for treating infections caused by Gram-negative multidrug-resistant bacteria. The mechanisms of mutational resistance to fosfomycin in Stenotrophomonas maltophilia, an opportunistic pathogen characterized by its low susceptibility to commonly used antibiotics, were studied in the current work. None of the mechanisms reported so far for other organisms, which include the production of fosfomycin-inactivating enzymes, target modification, induction of an alternative peptidoglycan biosynthesis pathway, and the impaired entry of the antibiotic, are involved in the acquisition of such resistance by this bacterial species. Instead, the unique cause of resistance in the mutants studied is the mutational inactivation of different enzymes belonging to the Embden-Meyerhof-Parnas central metabolism pathway. The amount of intracellular fosfomycin accumulation did not change in any of these mutants, showing that neither inactivation nor transport of the antibiotic is involved. Transcriptomic analysis also showed that the mutants did not present changes in the expression level of putative alternative peptidoglycan biosynthesis pathway genes or any related enzyme. Finally, the mutants did not present an increased phosphoenolpyruvate concentration that might compete with fosfomycin for its binding to MurA. On the basis of these results, we describe a completely novel mechanism of antibiotic resistance based on mutations of genes encoding metabolic enzymes.IMPORTANCE Antibiotic resistance has been largely considered a specific bacterial response to an antibiotic challenge. Indeed, its study has been mainly concentrated on mechanisms that affect the antibiotics (mutations in transporters, efflux pumps, and antibiotic-modifying enzymes, or their regulators) or their targets (i.e., target mutations, protection, or bypass). Usually, antibiotic resistance-associated metabolic changes were considered a consequence (fitness costs) and not a cause of antibiotic resistance. Herein, we show that alterations in the central carbon bacterial metabolism can also be the cause of antibiotic resistance. In the study presented here, Stenotrophomonas maltophilia acquires fosfomycin resistance through the inactivation of glycolytic enzymes belonging to the Embden-Meyerhof-Parnas pathway. Besides resistance to fosfomycin, this inactivation also impairs the bacterial gluconeogenic pathway. Together with previous work showing that antibiotic resistance can be under metabolic control, our results provide evidence that antibiotic resistance is intertwined with the bacterial metabolism.
Collapse
|
102
|
Buszewski B, Rogowska A, Railean-Plugaru V, Złoch M, Walczak-Skierska J, Pomastowski P. The Influence of Different Forms of Silver on Selected Pathogenic Bacteria. MATERIALS (BASEL, SWITZERLAND) 2020; 13:E2403. [PMID: 32456144 PMCID: PMC7287713 DOI: 10.3390/ma13102403] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 05/20/2020] [Accepted: 05/20/2020] [Indexed: 01/24/2023]
Abstract
The application of silver nanoparticles as an antibacterial agent is becoming more common. Unfortunately, their effect on microorganisms is still not fully understood. Therefore, this paper attempts to investigate the influence of silver ions, biologically synthesized silver nanoparticles and nanoparticles functionalized with antibiotics on molecular bacteria profiles. The initial stage of research was aimed at the mechanism determination involved in antibiotics sorption onto nanoparticles' surface. For this purpose, the kinetics study was performed. Next, the functionalized formulations were characterized by Fourier transform infrared spectroscopy (FT-IR), dynamic light scattering (DLS) and a zeta potential study. The results reveal that functionalization is a complex process, but does not significantly affect the stability of biocolloids. Furthermore, the antimicrobial assays, in most cases, have shown no increases in antibacterial activity after nanoparticle functionalization, which suggests that the functionalization process does not always generate the improved antimicrobial effect. Finally, the matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) technique was employed to characterize the changes in the molecular profile of bacteria treated with various antibacterial agents. The recorded spectra proved many differences in bacterial lipids and proteins profiles compared to untreated cells. In addition, the statistical analysis of recorded spectra revealed the strain-dependent nature of stress factors on the molecular profile of microorganisms.
Collapse
Affiliation(s)
- Bogusław Buszewski
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University in Torun, Wileńska 4, 87-100 Torun, Poland; (B.B.); (A.R.); (V.R.-P.); (M.Z.)
- Department of Environmental Chemistry and Bioanalytics, Faculty of Chemistry, Nicolaus Copernicus University in Torun, Gagarina 7, 87-100 Torun, Poland;
| | - Agnieszka Rogowska
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University in Torun, Wileńska 4, 87-100 Torun, Poland; (B.B.); (A.R.); (V.R.-P.); (M.Z.)
- Department of Environmental Chemistry and Bioanalytics, Faculty of Chemistry, Nicolaus Copernicus University in Torun, Gagarina 7, 87-100 Torun, Poland;
| | - Viorica Railean-Plugaru
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University in Torun, Wileńska 4, 87-100 Torun, Poland; (B.B.); (A.R.); (V.R.-P.); (M.Z.)
- Department of Environmental Chemistry and Bioanalytics, Faculty of Chemistry, Nicolaus Copernicus University in Torun, Gagarina 7, 87-100 Torun, Poland;
| | - Michał Złoch
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University in Torun, Wileńska 4, 87-100 Torun, Poland; (B.B.); (A.R.); (V.R.-P.); (M.Z.)
| | - Justyna Walczak-Skierska
- Department of Environmental Chemistry and Bioanalytics, Faculty of Chemistry, Nicolaus Copernicus University in Torun, Gagarina 7, 87-100 Torun, Poland;
| | - Paweł Pomastowski
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University in Torun, Wileńska 4, 87-100 Torun, Poland; (B.B.); (A.R.); (V.R.-P.); (M.Z.)
| |
Collapse
|
103
|
Gil-Gil T, Martínez JL, Blanco P. Mechanisms of antimicrobial resistance in Stenotrophomonas maltophilia: a review of current knowledge. Expert Rev Anti Infect Ther 2020; 18:335-347. [PMID: 32052662 DOI: 10.1080/14787210.2020.1730178] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: Stenotrophomonas maltophilia is a prototype of bacteria intrinsically resistant to antibiotics. The reduced susceptibility of this microorganism to antimicrobials mainly relies on the presence in its chromosome of genes encoding efflux pumps and antibiotic inactivating enzymes. Consequently, the therapeutic options for treating S. maltophilia infections are limited.Areas covered: Known mechanisms of intrinsic, acquired and phenotypic resistance to antibiotics of S. maltophilia and the consequences of such resistance for treating S. maltophilia infections are discussed. Acquisition of some genes, mainly those involved in co-trimoxazole resistance, contributes to acquired resistance. Mutation, mainly in the regulators of chromosomally-encoded antibiotic resistance genes, is a major cause for S. maltophilia acquisition of resistance. The expression of some of these genes is triggered by specific signals or stressors, which can lead to transient phenotypic resistance.Expert opinion: Treatment of S. maltophilia infections is difficult because this organism presents low susceptibility to antibiotics. Besides, it can acquire resistance to antimicrobials currently in use. Particularly problematic is the selection of mutants overexpressing efflux pumps since they present a multidrug resistance phenotype. The use of novel antimicrobials alone or in combination, together with the development of efflux pumps' inhibitors may help in fighting S. maltophilia infections.
Collapse
Affiliation(s)
| | | | - Paula Blanco
- Molecular Basis of Adaptation Laboratory, Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
104
|
Kim Y, Leung MHY, Kwok W, Fournié G, Li J, Lee PKH, Pfeiffer DU. Antibiotic resistance gene sharing networks and the effect of dietary nutritional content on the canine and feline gut resistome. Anim Microbiome 2020; 2:4. [PMID: 33500005 PMCID: PMC7807453 DOI: 10.1186/s42523-020-0022-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 01/29/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND As one of the most densely populated microbial communities on Earth, the gut microbiota serves as an important reservoir of antibiotic resistance genes (ARGs), referred to as the gut resistome. Here, we investigated the association of dietary nutritional content with gut ARG diversity and composition, using publicly available shotgun metagenomic sequence data generated from canine and feline fecal samples. Also, based on network theory, we explored ARG-sharing patterns between gut bacterial genera by identifying the linkage structure between metagenomic assemblies and their functional genes obtained from the same data. RESULTS In both canine and feline gut microbiota, an increase in protein and a reduction in carbohydrate in the diet were associated with increased ARG diversity. ARG diversity of the canine gut microbiota also increased, but less strongly, after a reduction in protein and an increase in carbohydrate in the diet. The association between ARG and taxonomic composition suggests that diet-induced changes in the gut microbiota may be responsible for changes in ARG composition, supporting the links between protein metabolism and antibiotic resistance in gut microbes. In the analysis of the ARG-sharing patterns, 22 ARGs were shared among 46 genera in the canine gut microbiota, and 11 ARGs among 28 genera in the feline gut microbiota. Of these ARGs, the tetracycline resistance gene tet(W) was shared among the largest number of genera, predominantly among Firmicutes genera. Bifidobacterium, a genus extensively used in the fermentation of dairy products and as probiotics, shared tet(W) with a wide variety of other genera. Finally, genera from the same phylum were more likely to share ARGs than with those from different phyla. CONCLUSIONS Our findings show that dietary nutritional content, especially protein content, is associated with the gut resistome and suggest future research to explore the impact of dietary intervention on the development of antibiotic resistance in clinically-relevant gut microbes. Our network analysis also reveals that the genetic composition of bacteria acts as an important barrier to the horizontal transfer of ARGs. By capturing the underlying gene-sharing relationships between different bacterial taxa from metagenomes, our network approach improves our understanding of horizontal gene transfer dynamics.
Collapse
Affiliation(s)
- Younjung Kim
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong, China.
| | - Marcus H Y Leung
- School of Energy and Environment, City University of Hong Kong, Hong Kong, China
| | - Wendy Kwok
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong, China
| | - Guillaume Fournié
- Department of Pathobiology and Population Sciences, Royal Veterinary College, London, UK
| | - Jun Li
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong, China.,School of Data Science, City University of Hong Kong, Hong Kong, China
| | - Patrick K H Lee
- School of Energy and Environment, City University of Hong Kong, Hong Kong, China
| | - Dirk U Pfeiffer
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong, China.,Department of Pathobiology and Population Sciences, Royal Veterinary College, London, UK
| |
Collapse
|
105
|
Hernando-Amado S, Sanz-García F, Martínez JL. Antibiotic Resistance Evolution Is Contingent on the Quorum-Sensing Response in Pseudomonas aeruginosa. Mol Biol Evol 2020; 36:2238-2251. [PMID: 31228244 DOI: 10.1093/molbev/msz144] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Different works have explored independently the evolution toward antibiotic resistance and the role of eco-adaptive mutations in the adaptation to a new habitat (as the infected host) of bacterial pathogens. However, knowledge about the connection between both processes is still limited. We address this issue by comparing the evolutionary trajectories toward antibiotic resistance of a Pseudomonas aeruginosa lasR defective mutant and its parental wild-type strain, when growing in presence of two ribosome-targeting antibiotics. Quorum-sensing lasR defective mutants are selected in P. aeruginosa populations causing chronic infections. Further, we observed they are also selected in vitro as a first adaptation for growing in culture medium. By using experimental evolution and whole-genome sequencing, we found that the evolutionary trajectories of P. aeruginosa in presence of these antibiotics are different in lasR defective and in wild-type backgrounds, both at the phenotypic and the genotypic levels. Recreation of a set of mutants in both genomic backgrounds (either wild type or lasR defective) allowed us to determine the existence of negative epistatic interactions between lasR and antibiotic resistance determinants. These epistatic interactions could lead to mutual contingency in the evolution of antibiotic resistance when P. aeruginosa colonizes a new habitat in presence of antibiotics. If lasR mutants are selected first, this would constraint antibiotic resistance evolution. Conversely, when resistance mutations (at least those studied in the present work) are selected, lasR mutants may not be selected in presence of antibiotics. These results underlie the importance of contingency and epistatic interactions in modulating antibiotic resistance evolution.
Collapse
|
106
|
Peng B, Li H, Peng X. Proteomics approach to understand bacterial antibiotic resistance strategies. Expert Rev Proteomics 2019; 16:829-839. [PMID: 31618606 DOI: 10.1080/14789450.2019.1681978] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Introduction: The understanding of novel antibiotic resistance mechanisms is essential to develop strategies against antibiotic-resistant pathogens, which has become an urgent task due to the worldwide emergence of antibiotic resistance. Areas covered: In this review, the authors summarize the recent progress on antibiotic resistance caused by lab-evolved bacteria and clinical multidrug-resistant bacterial pathogens from the proteomics perspective. Expert opinion: Proteomics provides a new platform for a comprehensive understanding of change in protein pathways that are engaged in antibiotics resistance, which is different from a genetic view that focuses on the role of an individual gene or protein. Further work is required to understand why and how the involved pathways are integrated for surviving antibiotic-mediated killing, to use other OMICs for better comprehension of antibiotic resistance mechanisms, and to develop reprogramming proteomics, which reverts an 'antibiotic resistance proteome' to an 'antibiotic sensitive or antibiotic sensitive-like' proteome, for the control of antibiotic-resistant pathogens.
Collapse
Affiliation(s)
- Bo Peng
- Center for Proteomics and Metabolomics, State Key Laboratory of Bio-Control, School of Life Sciences, Sun Yat-sen University , Guangzhou , People's Republic of China.,Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology , Qingdao , People's Republic of China.,Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai) , Zhuhai , People's Republic of China
| | - Hui Li
- Center for Proteomics and Metabolomics, State Key Laboratory of Bio-Control, School of Life Sciences, Sun Yat-sen University , Guangzhou , People's Republic of China.,Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology , Qingdao , People's Republic of China.,Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai) , Zhuhai , People's Republic of China
| | - Xuanxian Peng
- Center for Proteomics and Metabolomics, State Key Laboratory of Bio-Control, School of Life Sciences, Sun Yat-sen University , Guangzhou , People's Republic of China.,Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology , Qingdao , People's Republic of China.,Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai) , Zhuhai , People's Republic of China
| |
Collapse
|
107
|
Siebert C, Lindgren H, Ferré S, Villers C, Boisset S, Perard J, Sjöstedt A, Maurin M, Brochier-Armanet C, Couté Y, Renesto P. Francisella tularensis: FupA mutation contributes to fluoroquinolone resistance by increasing vesicle secretion and biofilm formation. Emerg Microbes Infect 2019; 8:808-822. [PMID: 31164053 PMCID: PMC6566608 DOI: 10.1080/22221751.2019.1615848] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Francisella tularensis is the causative agent in tularemia for which the high prevalence of treatment failure and relapse is a major concern. Directed-evolution experiments revealed that acquisition of fluoroquinolone (FQ) resistance was linked to factors in addition to mutations in DNA gyrase. Here, using F. tularensis live vaccine strain (LVS) as a model, we demonstrated that FupA/B (Fer-Utilization Protein) expression is linked to FQ susceptibility, and that the virulent strain F. tularensis subsp. tularensis SCHU S4 deleted for the homologous FupA protein exhibited even higher FQ resistance. In addition to an increased FQ minimal inhibitory concentration, LVSΔfupA/B displayed tolerance toward bactericidal compounds including ciprofloxacin and gentamicin. Interestingly, the FupA/B deletion was found to promote increased secretion of outer membrane vesicles (OMVs). Mass spectrometry-based quantitative proteomic characterization of vesicles from LVS and LVS∆fupA/B identified 801 proteins, including a subset of 23 proteins exhibiting differential abundance between both strains which may therefore contribute to the reduced antibiotic susceptibility of the FupA/B-deleted strain. We also demonstrated that OMVs are key structural elements of LVSΔfupA/B biofilms providing protection against FQ. These results provide a new basis for understanding and tackling antibiotic resistance and/or persistence of Francisella and other pathogenic members of the Thiotrichales class.
Collapse
Affiliation(s)
- Claire Siebert
- a TIMC-IMAG UMR 5525 - UGA CNRS , Grenoble , France.,b Centre National de Référence des Francisella , Centre Hospitalo-Universitaire Grenoble Alpes , Grenoble , France
| | - Helena Lindgren
- c Laboratory for Molecular Infection Medicine Sweden and Department of Clinical Microbiology , Umeå University , Umeå , Sweden
| | - Sabrina Ferré
- d Université Grenoble Alpes, CEA, Inserm, IRIG-BGE , Grenoble , France
| | - Corinne Villers
- a TIMC-IMAG UMR 5525 - UGA CNRS , Grenoble , France.,e Université de Caen Normandie, EA4655 U2RM , Caen , France
| | - Sandrine Boisset
- a TIMC-IMAG UMR 5525 - UGA CNRS , Grenoble , France.,b Centre National de Référence des Francisella , Centre Hospitalo-Universitaire Grenoble Alpes , Grenoble , France
| | - Julien Perard
- f Université Grenoble Alpes, CNRS, CEA, BIG-LCBM , Grenoble , France
| | - Anders Sjöstedt
- c Laboratory for Molecular Infection Medicine Sweden and Department of Clinical Microbiology , Umeå University , Umeå , Sweden
| | - Max Maurin
- a TIMC-IMAG UMR 5525 - UGA CNRS , Grenoble , France.,b Centre National de Référence des Francisella , Centre Hospitalo-Universitaire Grenoble Alpes , Grenoble , France
| | - Céline Brochier-Armanet
- g Laboratoire de Biométrie et Biologie Évolutive , Université Claude Bernard Lyon 1, CNRS, UMR5558 , Villeurbanne , France
| | - Yohann Couté
- d Université Grenoble Alpes, CEA, Inserm, IRIG-BGE , Grenoble , France
| | | |
Collapse
|
108
|
Lin W, Wan K, Zeng J, Li J, Li X, Yu X. Low nutrient levels as drinking water conditions can reduce the fitness cost of efflux pump-mediated ciprofloxacin resistance in Pseudomonas aeruginosa. J Environ Sci (China) 2019; 83:123-132. [PMID: 31221375 DOI: 10.1016/j.jes.2019.03.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 03/26/2019] [Accepted: 03/26/2019] [Indexed: 06/09/2023]
Abstract
The long-term persistence of antibiotic resistance in the environment, especially in drinking water, is a public health concern. Expression of an efflux pump, an important mechanism of resistance to antibiotics, usually confers a fitness cost in bacteria. In this study, we aimed to determine why antibiotic resistance conferred by overexpression of an efflux pump persisted in low-nutrient environments (TOC < 10 mg/L) such as drinking and source water in which antibiotic selective pressure might be very low or even absent. Competition experiments between wild-type Pseudomonas aeruginosa and ciprofloxacin-resistant mutants revealed that the fitness cost of ciprofloxacin resistance significantly decreased (p < 0.05) under low-nutrient (0.5 mg/L total organic carbon (TOC)) relative to high-nutrient (500 mg/L TOC) conditions. Mechanisms underlying this fitness cost were analyzed. The mexD gene expression in resistant bacteria (cip_3 strain) was significantly lower (p < 0.05) in low-nutrient conditions, with 10 mg/L TOC ((8.01 ± 0.82)-fold), than in high-nutrient conditions, with 500 mg/L TOC ((48.89 ± 4.16)-fold). Moreover, rpoS gene expression in resistant bacteria ((1.36 ± 0.13)-fold) was significantly lower (p < 0.05) than that in the wild-type strain ((2.78 ± 0.29)-fold) under low-nutrient conditions (10 mg/L TOC), suggesting a growth advantage. Furthermore, the difference in metabolic activity between the two competing strains was significantly smaller (p < 0.05) in low-nutrient conditions (5 and 0.5 mg/L TOC). These results suggest that nutrient levels are a key factor in determining the persistence of antibiotic resistance conferred by efflux pumps in the natural environment with trace amounts or no antibiotics.
Collapse
Affiliation(s)
- Wenfang Lin
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
| | - Kun Wan
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jie Zeng
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jingjing Li
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
| | - Xi Li
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xin Yu
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China.
| |
Collapse
|
109
|
Stokes JM, Lopatkin AJ, Lobritz MA, Collins JJ. Bacterial Metabolism and Antibiotic Efficacy. Cell Metab 2019; 30:251-259. [PMID: 31279676 PMCID: PMC6990394 DOI: 10.1016/j.cmet.2019.06.009] [Citation(s) in RCA: 361] [Impact Index Per Article: 60.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/06/2019] [Accepted: 06/10/2019] [Indexed: 02/07/2023]
Abstract
Antibiotics target energy-consuming processes. As such, perturbations to bacterial metabolic homeostasis are significant consequences of treatment. Here, we describe three postulates that collectively define antibiotic efficacy in the context of bacterial metabolism: (1) antibiotics alter the metabolic state of bacteria, which contributes to the resulting death or stasis; (2) the metabolic state of bacteria influences their susceptibility to antibiotics; and (3) antibiotic efficacy can be enhanced by altering the metabolic state of bacteria. Altogether, we aim to emphasize the close relationship between bacterial metabolism and antibiotic efficacy as well as propose areas of exploration to develop novel antibiotics that optimally exploit bacterial metabolic networks.
Collapse
Affiliation(s)
- Jonathan M Stokes
- Institute for Medical Engineering & Science, Department of Biological Engineering, and Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Infectious Disease & Microbiome Program, Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA; Machine Learning for Pharmaceutical Discovery and Synthesis Consortium, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Allison J Lopatkin
- Institute for Medical Engineering & Science, Department of Biological Engineering, and Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Infectious Disease & Microbiome Program, Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Michael A Lobritz
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, 4070 Basel, Switzerland
| | - James J Collins
- Institute for Medical Engineering & Science, Department of Biological Engineering, and Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Infectious Disease & Microbiome Program, Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA; Harvard-MIT Program in Health Sciences and Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
110
|
Matteoli FP, Passarelli-Araujo H, Pedrosa-Silva F, Olivares FL, Venancio TM. Population structure and pangenome analysis of Enterobacter bugandensis uncover the presence of bla CTX-M-55, bla NDM-5 and bla IMI-1, along with sophisticated iron acquisition strategies. Genomics 2019; 112:1182-1191. [PMID: 31279858 DOI: 10.1016/j.ygeno.2019.07.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/02/2019] [Accepted: 07/03/2019] [Indexed: 01/22/2023]
Abstract
Enterobacter bugandensis is a recently described species that has been largely associated with nosocomial infections. We report the genome of a non-clinical E. bugandensis strain, which was integrated with publicly available genomes to study the pangenome and general population structure of E. bugandensis. Core- and whole-genome multilocus sequence typing allowed the detection of five E. bugandensis phylogroups (PG-A to E), which contain important antimicrobial resistance and virulence determinants. We uncovered several extended-spectrum β-lactamases, including blaCTX-M-55 and blaNDM-5, present in an IncX replicon type plasmid, described here for the first time in E. bugandensis. Genetic context analysis of blaNDM-5 revealed the resemblance of this plasmid with other IncX plasmids from other bacteria from the same country. Three distinctive siderophore producing operons were found in E. bugandensis: enterobactin (ent), aerobactin (iuc/iut), and salmochelin (iro). Our findings provide novel insights on the lifestyle, physiology, antimicrobial, and virulence profiles of E. bugandensis.
Collapse
Affiliation(s)
- Filipe P Matteoli
- Laboratório de Química e Função de Proteínas e Peptídeos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro (UENF), Brazil
| | - Hemanoel Passarelli-Araujo
- Laboratório de Química e Função de Proteínas e Peptídeos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro (UENF), Brazil; Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Francisnei Pedrosa-Silva
- Laboratório de Química e Função de Proteínas e Peptídeos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro (UENF), Brazil
| | - Fabio L Olivares
- Núcleo de Desenvolvimento de Insumos Biológicos para a Agricultura (NUDIBA), UENF, Brazil; Laboratório de Biologia Celular e Tecidual, Centro de Biociências e Biotecnologia, UENF, Brazil
| | - Thiago M Venancio
- Laboratório de Química e Função de Proteínas e Peptídeos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro (UENF), Brazil.
| |
Collapse
|
111
|
Danner MC, Robertson A, Behrends V, Reiss J. Antibiotic pollution in surface fresh waters: Occurrence and effects. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 664:793-804. [PMID: 30763859 DOI: 10.1016/j.scitotenv.2019.01.406] [Citation(s) in RCA: 457] [Impact Index Per Article: 76.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 01/25/2019] [Accepted: 01/30/2019] [Indexed: 05/25/2023]
Abstract
Worldwide, antibiotic usage exceeds 100,000 tons per year and there is increasing concern over the fate of these substances. Antibiotics are ubiquitous in the environment and significant concentrations have been detected in fresh waters. In this review, we highlight important aspects of antibiotic pollution in fresh waters: that concentrations of antibiotics in the environment are substantial, that micro-organisms are susceptible to this, that bacteria can evolve resistance in the environment, and that antibiotic pollution affects natural food webs while interacting with other stressors; which taken together poses a number of challenges for environmental scientists. In the literature, we found examples of considerable antibiotic pollution in fresh waters. In the Americas, antibiotic concentrations of up to 15 μg/L have been measured; with higher concentrations reported from European and African studies (over 10 μg/L and 50 μg/L respectively), and in Asian-pacific countries concentrations over 450 μg/L have been detected. While these concentrations might not be deemed harmful to humans, non-target freshwater organisms could be affected by them. Bioassays show that some of the antibiotics found in surface waters affect microbes at concentrations below 10 μg/L. Among the most potent antibiotics are those that prevail in streams and rivers in these concentrations, such as ciprofloxacin. Sub-lethal concentrations might not kill prokaryotes but contribute to increased bacterial resistance and change the composition of single-celled communities, as demonstrated in laboratory experiments. This has implications for the microbial food web (e.g. interactions among and between bacteria and their protozoan consumers) and by extension, larger organisms and ecosystem health. The fact that the effects of antibiotics are extremely context-dependent represents a challenge, particularly for in vitro research. We suggest future research avenues, taking into account food web experiments, antibiotics interacting with one another (and other stressors) and discuss how these can help to answer multi-layered research questions.
Collapse
Affiliation(s)
- Marie-Claire Danner
- Department of Life Sciences, Whitelands College, Roehampton University, London SW15 4JD, United Kingdom.
| | - Anne Robertson
- Department of Life Sciences, Whitelands College, Roehampton University, London SW15 4JD, United Kingdom
| | - Volker Behrends
- Department of Life Sciences, Whitelands College, Roehampton University, London SW15 4JD, United Kingdom
| | - Julia Reiss
- Department of Life Sciences, Whitelands College, Roehampton University, London SW15 4JD, United Kingdom
| |
Collapse
|
112
|
Poudel S, Tsunemoto H, Meehan M, Szubin R, Olson CA, Lamsa A, Seif Y, Dillon N, Vrbanac A, Sugie J, Dahesh S, Monk JM, Dorrestein PC, Pogliano J, Knight R, Nizet V, Palsson BO, Feist AM. Characterization of CA-MRSA TCH1516 exposed to nafcillin in bacteriological and physiological media. Sci Data 2019; 6:43. [PMID: 31028276 PMCID: PMC6486602 DOI: 10.1038/s41597-019-0051-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 03/07/2019] [Indexed: 12/05/2022] Open
Abstract
Cation adjusted-Mueller Hinton Broth (CA-MHB) is the standard bacteriological medium utilized in the clinic for the determination of antibiotic susceptibility. However, a growing number of literature has demonstrated that media conditions can cause a substantial difference in the efficacy of antibiotics and antimicrobials. Recent studies have also shown that minimum inhibitory concentration (MIC) tests performed in standard cell culture media (e.g. RPMI and DMEM) are more indicative of in vivo antibiotic efficacy, presumably because they are a better proxy for the human host’s physiological conditions. The basis for the bacterial media dependent susceptibility to antibiotics remains undefined. To address this question, we characterized the physiological response of methicillin-resistant Staphylococcus aureus (MRSA) during exposure to sub-inhibitory concentrations of the beta-lactam antibiotic nafcillin in either CA-MHB or RPMI + 10% LB (R10LB). Here, we present high quality transcriptomic, exo-metabolomic and morphological data paired with growth and susceptibility results for MRSA cultured in either standard bacteriologic or more physiologic relevant medium. Design Type(s) | replicate design • transcription profiling design • sequence analysis objective | Measurement Type(s) | transcription profiling assay • cellular morphology • exo-metabolome • growth | Technology Type(s) | RNA sequencing • fluorescence microscopy • liquid chromatography-tandem mass spectrometry • high performance liquid chromatography • Optical Density Measurement | Factor Type(s) | culture medium • biological replicate • experimental condition | Sample Characteristic(s) | Staphylococcus aureus • culturing environment |
Machine-accessible metadata file describing the reported data (ISA-Tab format)
Collapse
Affiliation(s)
- Saugat Poudel
- Department of Bioengineering, University of California, San Diego, La Jolla, USA
| | - Hannah Tsunemoto
- Division of Biological Sciences, University of California San Diego, La Jolla, CA, 92093, USA
| | - Michael Meehan
- Collaborative Mass Spectrometry Innovation Center, University of California, San Diego, La Jolla, California, USA.,Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Richard Szubin
- Department of Bioengineering, University of California, San Diego, La Jolla, USA
| | - Connor A Olson
- Department of Bioengineering, University of California, San Diego, La Jolla, USA
| | - Anne Lamsa
- Division of Biological Sciences, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yara Seif
- Department of Bioengineering, University of California, San Diego, La Jolla, USA
| | - Nicholas Dillon
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Alison Vrbanac
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Joseph Sugie
- Division of Biological Sciences, University of California San Diego, La Jolla, CA, 92093, USA
| | - Samira Dahesh
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Jonathan M Monk
- Department of Bioengineering, University of California, San Diego, La Jolla, USA
| | - Pieter C Dorrestein
- Collaborative Mass Spectrometry Innovation Center, University of California, San Diego, La Jolla, California, USA.,Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA.,Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA, 92093, USA.,Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, 92093, USA
| | - Joseph Pogliano
- Division of Biological Sciences, University of California San Diego, La Jolla, CA, 92093, USA
| | - Rob Knight
- Department of Bioengineering, University of California, San Diego, La Jolla, USA.,Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA.,Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA, 92093, USA.,Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, 92093, USA
| | - Victor Nizet
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA.,Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA.,Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, 92093, USA
| | - Bernhard O Palsson
- Department of Bioengineering, University of California, San Diego, La Jolla, USA.,Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA.,Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet, Building 220, 2800, Kongens, Lyngby, Denmark.,Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, 92093, USA
| | - Adam M Feist
- Department of Bioengineering, University of California, San Diego, La Jolla, USA. .,Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet, Building 220, 2800, Kongens, Lyngby, Denmark.
| |
Collapse
|
113
|
Hashemi MM, Holden BS, Coburn J, Taylor MF, Weber S, Hilton B, Zaugg AL, McEwan C, Carson R, Andersen JL, Price JC, Deng S, Savage PB. Proteomic Analysis of Resistance of Gram-Negative Bacteria to Chlorhexidine and Impacts on Susceptibility to Colistin, Antimicrobial Peptides, and Ceragenins. Front Microbiol 2019; 10:210. [PMID: 30833936 PMCID: PMC6388577 DOI: 10.3389/fmicb.2019.00210] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 01/24/2019] [Indexed: 12/19/2022] Open
Abstract
Use of chlorhexidine in clinical settings has led to concerns that repeated exposure of bacteria to sub-lethal doses of chlorhexidine might result in chlorhexidine resistance and cross resistance with other cationic antimicrobials including colistin, endogenous antimicrobial peptides (AMPs) and their mimics, ceragenins. We have previously shown that colistin-resistant Gram-negative bacteria remain susceptible to AMPs and ceragenins. Here, we investigated the potential for cross resistance between chlorhexidine, colistin, AMPs and ceragenins by serial exposure of standard strains of Gram-negative bacteria to chlorhexidine to generate resistant populations of organisms. Furthermore, we performed a proteomics study on the chlorhexidine-resistant strains and compared them to the wild-type strains to find the pathways by which bacteria develop resistance to chlorhexidine. Serial exposure of Gram-negative bacteria to chlorhexidine resulted in four- to eight-fold increases in minimum inhibitory concentrations (MICs). Chlorhexidine-resistant organisms showed decreased susceptibility to colistin (8- to 32-fold increases in MICs) despite not being exposed to colistin. In contrast, chlorhexidine-resistant organisms had the same MICs as the original strains when tested with representative AMPs (LL-37 and magainin I) and ceragenins (CSA-44 and CSA-131). These results imply that there may be a connection between the emergence of highly colistin-resistant Gram-negative pathogens and the prevalence of chlorhexidine usage. Yet, use of chlorhexidine may not impact innate immune defenses (e.g., AMPs) and their mimics (e.g., ceragenins). Here, we also show that chlorhexidine resistance is associated with upregulation of proteins involved in the assembly of LPS for outer membrane biogenesis and virulence factors in Pseudomonas aeruginosa. Additionally, resistance to chlorhexidine resulted in elevated expression levels of proteins associated with chaperones, efflux pumps, flagella and cell metabolism. This study provides a comprehensive overview of the evolutionary proteomic changes in P. aeruginosa following exposure to chlorhexidine and colistin. These results have important clinical implications considering the continuous application of chlorhexidine in hospitals that could influence the emergence of colistin-resistant strains.
Collapse
Affiliation(s)
- Marjan M Hashemi
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, United States
| | - Brett S Holden
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, United States
| | - Jordan Coburn
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, United States
| | - Maddison F Taylor
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, United States
| | - Scott Weber
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, United States
| | - Brian Hilton
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, United States
| | - Aaron L Zaugg
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, United States
| | - Colten McEwan
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, United States
| | - Richard Carson
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, United States
| | - Joshua L Andersen
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, United States
| | - John C Price
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, United States
| | - Shenglou Deng
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, United States
| | - Paul B Savage
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, United States
| |
Collapse
|
114
|
Banerjee D, Raghunathan A. Constraints-based analysis identifies NAD+ recycling through metabolic reprogramming in antibiotic resistant Chromobacterium violaceum. PLoS One 2019; 14:e0210008. [PMID: 30608971 PMCID: PMC6319732 DOI: 10.1371/journal.pone.0210008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 12/14/2018] [Indexed: 12/15/2022] Open
Abstract
In the post genomic era, high throughput data augment stoichiometric flux balance models to compute accurate metabolic flux states, growth and energy phenotypes. Investigating altered metabolism in the context of evolved resistant genotypes potentially provide simple strategies to overcome drug resistance and induce susceptibility to existing antibiotics. A genome-scale metabolic model (GSMM) for Chromobacterium violaceum, an opportunistic human pathogen, was reconstructed using legacy data. Experimental constraints were used to represent antibiotic susceptible and resistant populations. Model predictions were validated using growth and respiration data successfully. Differential flux distribution and metabolic reprogramming were identified as a response to antibiotics, chloramphenicol and streptomycin. Streptomycin resistant populations (StrpR) redirected tricarboxylic acid (TCA) cycle flux through the glyoxylate shunt. Chloramphenicol resistant populations (ChlR) resorted to overflow metabolism producing acetate and formate. This switch to fermentative metabolism is potentially through excess reducing equivalents and increased NADH/NAD ratios. Reduced proton gradients and changed Proton Motive Force (PMF) induced by antibiotics were also predicted and verified experimentally using flow cytometry based membrane potential measurements. Pareto analysis of NADH and ATP maintenance showed the decoupling of electron transfer and ATP synthesis in StrpR. Redox homeostasis and NAD+ cycling through rewiring metabolic flux was implicated in re-sensitizing antibiotic resistant C. violaceum. These approaches can be used to probe metabolic vulnerabilities of resistant pathogens. On the verge of a post-antibiotic era, we foresee a critical need for systems level understanding of pathogens and host interaction to extend shelf life of antibiotics and strategize novel therapies.
Collapse
Affiliation(s)
- Deepanwita Banerjee
- Chemical Engineering Division, CSIR-National Chemical Laboratory, Pune, Maharashtra, India
| | - Anu Raghunathan
- Chemical Engineering Division, CSIR-National Chemical Laboratory, Pune, Maharashtra, India
| |
Collapse
|
115
|
Dunphy LJ, Yen P, Papin JA. Integrated Experimental and Computational Analyses Reveal Differential Metabolic Functionality in Antibiotic-Resistant Pseudomonas aeruginosa. Cell Syst 2019; 8:3-14.e3. [PMID: 30611675 DOI: 10.1016/j.cels.2018.12.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 10/08/2018] [Accepted: 12/04/2018] [Indexed: 12/13/2022]
Abstract
Metabolic adaptations accompanying the development of antibiotic resistance in bacteria remain poorly understood. To study this relationship, we profiled the growth of lab-evolved antibiotic-resistant lineages of the opportunistic pathogen Pseudomonas aeruginosa across 190 unique carbon sources. Our data revealed that the evolution of antibiotic resistance resulted in systems-level changes to growth dynamics and metabolic phenotype. A genome-scale metabolic network reconstruction of P. aeruginosa was paired with whole-genome sequencing data to predict genes contributing to observed changes in metabolism. We experimentally validated computational predictions to identify mutations in resistant P. aeruginosa affecting loss of catabolic function. Finally, we found a shared metabolic phenotype between lab-evolved P. aeruginosa and clinical isolates with similar mutational landscapes. Our results build upon previous knowledge of antibiotic-induced metabolic adaptation and provide a framework for the identification of metabolic limitations in antibiotic-resistant pathogens.
Collapse
Affiliation(s)
- Laura J Dunphy
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Phillip Yen
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Jason A Papin
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA; Department of Medicine, Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA; Department of Biochemistry & Molecular Genetics, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
116
|
Cokol M, Li C, Chandrasekaran S. Chemogenomic model identifies synergistic drug combinations robust to the pathogen microenvironment. PLoS Comput Biol 2018; 14:e1006677. [PMID: 30596642 PMCID: PMC6329523 DOI: 10.1371/journal.pcbi.1006677] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 01/11/2019] [Accepted: 11/27/2018] [Indexed: 01/31/2023] Open
Abstract
Antibiotics need to be effective in diverse environments in vivo. However, the pathogen microenvironment can have a significant impact on antibiotic potency. Further, antibiotics are increasingly used in combinations to combat resistance, yet, the effect of microenvironments on drug-combination efficacy is unknown. To exhaustively explore the impact of diverse microenvironments on drug-combinations, here we develop a computational framework—Metabolism And GENomics-based Tailoring of Antibiotic regimens (MAGENTA). MAGENTA uses chemogenomic profiles of individual drugs and metabolic perturbations to predict synergistic or antagonistic drug-interactions in different microenvironments. We uncovered antibiotic combinations with robust synergy across nine distinct environments against both E. coli and A. baumannii by searching through 2556 drug-combinations of 72 drugs. MAGENTA also accurately predicted the change in efficacy of bacteriostatic and bactericidal drug-combinations during growth in glycerol media, which we confirmed experimentally in both microbes. Our approach identified genes in glycolysis and glyoxylate pathway as top predictors of synergy and antagonism respectively. Our systems approach enables tailoring of antibiotic therapies based on the pathogen microenvironment. The antibiotic resistance epidemic has created a pressing need to understand factors that influence antibiotic efficacy. An often-overlooked factor in the search for new treatments is the pathogen environment. Understanding the differences in pathogen sensitivity to antibiotics in lab conditions versus inside the host is necessary for translating new discoveries into the clinic. Hence, we experimentally measured the sensitivity of E. coli to drugs and drug combinations in different metabolic conditions. Our data revealed that the environment dramatically changes treatment potency. Each antibiotic class was affected uniquely by each metabolic condition. The large number of metabolic conditions inside the host greatly complicates the identification of effective therapies. To address this challenge, we present a computational approach called MAGENTA that accurately predicted efficacy of antibiotic regimens in different conditions, which we confirmed experimentally. Furthermore, we show that MAGENTA can be applied to other bacterial pathogens such as A. baumannii and M. tuberculosis without the need for generating expensive data in each organism. MAGENTA accurately predicted efficacy in the pathogen A. baumannii using data from E. coli by identifying genes that are common between the two bacteria. Our study revealed the significant yet predictable impact of environment on drug combination potency.
Collapse
Affiliation(s)
- Murat Cokol
- Axcella Health, Cambridge, Massachusetts, United States of America
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, Massachusetts, United States of America
- Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul, Turkey
- * E-mail: (SC); (MC)
| | - Chen Li
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Sriram Chandrasekaran
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail: (SC); (MC)
| |
Collapse
|
117
|
Zhao X, Chen H, Jin Z, Li L, Zhang J, Kong X. GC-MS-based metabolomics analysis reveals L-aspartate enhances the antibiotic sensitivity of neomycin sulfate-resistant Aeromonas hydrophila. JOURNAL OF FISH DISEASES 2018; 41:1831-1841. [PMID: 30259527 DOI: 10.1111/jfd.12894] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 06/23/2018] [Accepted: 06/25/2018] [Indexed: 06/08/2023]
Abstract
Neomycin sulfate, a kind of drug known as aminoglycoside antibiotic, can be used to treat external or internal bacterial infections. However, there are increasing signs that antibiotics use in aquaculture may have a long-term and permanent potential to select for antibiotic-resistant bacteria in the aquatic environment. In the present study, we aimed to identify key metabolic biomarker whose abundance is correlated with neomycin sulfate resistance in A. hydrophila by gas chromatography-mass spectrometry (GC-MS). Through bioinformatics analysis, L-aspartate was identified as the most crucial biomarker in neomycin sulfate-resistant A. hydrophila and a potential modulator of neomycin sulfate resistance. It was validated as a synergist that incubation of neomycin sulfate-susceptible or -resistant A. hydrophila with exogenous L-aspartate sensitized the bacteria to neomycin sulfate in vitro assay. Moreover, L-aspartate also significantly improved the bactericidal efficacy of neomycin sulfate in Carassius auratus, and increased the survival rate of fish after A. hydrophila challenge. This study presents a novel approach in fighting against antibiotic-resistant pathogens through exploration of antibiotic-resistant biomarkers.
Collapse
Affiliation(s)
- Xianliang Zhao
- College of Fisheries, Henan Normal University, Xinxiang, China
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China
| | - He Chen
- College of Fisheries, Henan Normal University, Xinxiang, China
| | - Zhaohui Jin
- College of Fisheries, Henan Normal University, Xinxiang, China
| | - Li Li
- College of Fisheries, Henan Normal University, Xinxiang, China
| | - Jie Zhang
- College of Fisheries, Henan Normal University, Xinxiang, China
| | - Xianghui Kong
- College of Fisheries, Henan Normal University, Xinxiang, China
| |
Collapse
|
118
|
Borkowski A, Gutowski Ł, Syczewski M, Cłapa T, Czerwonka G. Adaptation of bacteria Escherichia coli in presence of quaternary ammonium ionic liquids. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2018; 164:370-378. [PMID: 30138820 DOI: 10.1016/j.ecoenv.2018.08.048] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 08/08/2018] [Accepted: 08/14/2018] [Indexed: 06/08/2023]
Abstract
This paper presents the adaptation of Escherichia coli Gram-negative bacteria to increased concentrations of ionic liquids. Theophylline-based quaternary ammonium salts were used as an example of an ionic liquid that on the one hand includes an anion of natural origin and on the other hand is characterized by amphiphilic properties due to aliphatic chains in its structure. Theophylline-based ionic liquids can be synthesized relatively cheaply and easily and can exhibit strong antibacterial properties depending on the alkyl chain length. These compounds can also strongly affect bacterial membrane properties, including changes in electrokinetic potential as well as net surface charge. The experiments performed in this study succeeded in obtaining bacterial cultures growing at a tetradecyltrimethylammonium theophyllinate concentration three times higher than the minimum inhibition and bactericidal concentration. The adapted bacteria were characterized by intriguing changes in morphology and grew in the form of almost one-millimeter spheres in a liquid medium. It was shown that cultivation of adapted bacteria with tetradecyltrimethylammonium theophyllinate resulted in changes in the lipid membrane composition and protein patterns of the bacterial lysates, depending on the ionic liquid concentration. This study also revealed that such bacterial adaptation can increase sensitivity to antibiotics by affecting membrane properties like ionophores. These results can be potentially important with regard to synergistic or antagonistic action with other bactericidal compounds like antibiotics and nanoparticles.
Collapse
Affiliation(s)
- Andrzej Borkowski
- Faculty of Geology, Geomicrobiology Laboratory, University of Warsaw, Żwirki i Wigury 93, 02-089 Warsaw, Poland.
| | - Łukasz Gutowski
- Faculty of Advanced Technologies and Chemistry, Military University of Technology, Urbanowicza 2, 00-908 Warsaw, Poland
| | - Marcin Syczewski
- Faculty of Geology, Geomicrobiology Laboratory, University of Warsaw, Żwirki i Wigury 93, 02-089 Warsaw, Poland
| | - Tomasz Cłapa
- Department of General and Environmental Microbiology, Poznan University of Life Sciences, Szydłowska 50, 60-656 Poznan, Poland
| | - Grzegorz Czerwonka
- Department of Microbiology, Institute of Biology, Jan Kochanowski University in Kielce, Świętokrzyska 15, 25-406 Kielce, Poland
| |
Collapse
|
119
|
Pusic P, Sonnleitner E, Krennmayr B, Heitzinger DA, Wolfinger MT, Resch A, Bläsi U. Harnessing Metabolic Regulation to Increase Hfq-Dependent Antibiotic Susceptibility in Pseudomonas aeruginosa. Front Microbiol 2018; 9:2709. [PMID: 30473687 PMCID: PMC6237836 DOI: 10.3389/fmicb.2018.02709] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 10/23/2018] [Indexed: 01/04/2023] Open
Abstract
The opportunistic human pathogen Pseudomonas aeruginosa is responsible for ~ 10% of hospital-acquired infections worldwide. It is notorious for its high level resistance toward many antibiotics, and the number of multi-drug resistant clinical isolates is steadily increasing. A better understanding of the molecular mechanisms underlying drug resistance is crucial for the development of novel antimicrobials and alternative strategies such as enhanced sensitization of bacteria to antibiotics in use. In P. aeruginosa several uptake channels for amino-acids and carbon sources can serve simultaneously as entry ports for antibiotics. The respective genes are often controlled by carbon catabolite repression (CCR). We have recently shown that Hfq in concert with Crc acts as a translational repressor during CCR. This function is counteracted by the regulatory RNA CrcZ, which functions as a decoy to abrogate Hfq-mediated translational repression of catabolic genes. Here, we report an increased susceptibility of P. aeruginosa hfq deletion strains to different classes of antibiotics. Transcriptome analyses indicated that Hfq impacts on different mechanisms known to be involved in antibiotic susceptibility, viz import and efflux, energy metabolism, cell wall and LPS composition as well as on the c-di-GMP levels. Furthermore, we show that sequestration of Hfq by CrcZ, which was over-produced or induced by non-preferred carbon-sources, enhances the sensitivity toward antibiotics. Thus, controlled synthesis of CrcZ could provide a means to (re)sensitize P. aeruginosa to different classes of antibiotics.
Collapse
Affiliation(s)
- Petra Pusic
- Max F. Perutz Laboratories, Department of Microbiology, Immunobiology and Genetics, Vienna Biocenter, University of Vienna, Vienna, Austria
| | - Elisabeth Sonnleitner
- Max F. Perutz Laboratories, Department of Microbiology, Immunobiology and Genetics, Vienna Biocenter, University of Vienna, Vienna, Austria
| | - Beatrice Krennmayr
- Max F. Perutz Laboratories, Department of Microbiology, Immunobiology and Genetics, Vienna Biocenter, University of Vienna, Vienna, Austria
| | - Dorothea A. Heitzinger
- Max F. Perutz Laboratories, Department of Microbiology, Immunobiology and Genetics, Vienna Biocenter, University of Vienna, Vienna, Austria
| | | | - Armin Resch
- Max F. Perutz Laboratories, Department of Microbiology, Immunobiology and Genetics, Vienna Biocenter, University of Vienna, Vienna, Austria
| | - Udo Bläsi
- Max F. Perutz Laboratories, Department of Microbiology, Immunobiology and Genetics, Vienna Biocenter, University of Vienna, Vienna, Austria
| |
Collapse
|
120
|
Sanz-García F, Hernando-Amado S, Martínez JL. Mutational Evolution of Pseudomonas aeruginosa Resistance to Ribosome-Targeting Antibiotics. Front Genet 2018; 9:451. [PMID: 30405685 PMCID: PMC6200844 DOI: 10.3389/fgene.2018.00451] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 09/18/2018] [Indexed: 01/21/2023] Open
Abstract
The present work examines the evolutionary trajectories of replicate Pseudomonas aeruginosa cultures in presence of the ribosome-targeting antibiotics tobramycin and tigecycline. It is known that large number of mutations across different genes - and therefore a large number of potential pathways - may be involved in resistance to any single antibiotic. Thus, evolution toward resistance might, to a large degree, rely on stochasticity, which might preclude the use of predictive strategies for fighting antibiotic resistance. However, the present results show that P. aeruginosa populations evolving in parallel in the presence of antibiotics (either tobramycin or tigecycline) follow a set of trajectories that present common elements. In addition, the pattern of resistance mutations involved include common elements for these two ribosome-targeting antimicrobials. This indicates that mutational evolution toward resistance (and perhaps other properties) is to a certain degree deterministic and, consequently, predictable. These findings are of interest, not just for P. aeruginosa, but in understanding the general rules involved in the evolution of antibiotic resistance also. In addition, the results indicate that bacteria can evolve toward higher levels of resistance to antibiotics against which they are considered to be intrinsically resistant, as tigecycline in the case of P. aeruginosa and that this may confer cross-resistance to other antibiotics of therapeutic value. Our results are particularly relevant in the case of patients under empiric treatment with tigecycline, which frequently suffer P. aeruginosa superinfections.
Collapse
Affiliation(s)
| | - Sara Hernando-Amado
- Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - José L. Martínez
- Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| |
Collapse
|
121
|
Patkari M, Kumbhar C, Nag A, Mehra S. Distinct transcriptomic response of S. coelicolor to ciprofloxacin in a nutrient-rich environment. Appl Microbiol Biotechnol 2018; 102:10623-10643. [PMID: 30327831 DOI: 10.1007/s00253-018-9398-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 09/13/2018] [Accepted: 09/14/2018] [Indexed: 02/07/2023]
Abstract
With the rising threat of anti-microbial resistance (AMR), there is an urgent need to enhance efficacy of existing antibiotics. Understanding the myriad mechanisms through which bacteria evade these drugs would be of immense value to designing novel strategies against them. Streptomyces coelicolor A3(2) M145 belongs to the actinomyctes species that are responsible for more than two-thirds of antibiotics. This group of bacteria therefore encodes for various mechanisms that can resist both endogenous and non-endogenous antibiotics. In an earlier study, we had studied the transcriptomic response of these bacteria to ciprofloxacin, when cultured in a minimal media. In this work, we investigate why the minimum inhibitory concentration of the drug increases by fourfold when the bacteria are grown in a nutrient-rich media. Through transcriptomic, biochemical, and microscopic studies, we show that S. coelicolor responds to ciprofloxacin in a concentration-dependent manner. While, sub-inhibitory concentration of the drug primarily causes oxidative stress, the inhibitory concentration of ciprofloxacin evokes a more severe genome-wide response in the cell, which ranges from the familiar upregulation of the SOS response and DNA repair pathways to the widespread alterations in the central metabolism pathway to accommodate the increased needs of nucleotides and other precursors. Further, the upregulation of peptidoglycan synthesis genes, along with microscopy images, suggest alterations in the cell morphology to increase fitness of the bacteria during the antibiotic stress. The data also points to the enhanced efflux activity in cells cultured in rich media that contributes significantly towards reducing intracellular drug concentration and thus promotes survival.
Collapse
Affiliation(s)
- Minal Patkari
- Department of Chemical Engineering, IIT Bombay, Powai, 400076, India
- Department of Civil and Environmental Engineering, University of North Carolina at Charlotte, Charlotte, NC, USA
| | | | - Ankita Nag
- Department of Chemical Engineering, IIT Bombay, Powai, 400076, India
| | - Sarika Mehra
- Department of Chemical Engineering, IIT Bombay, Powai, 400076, India.
| |
Collapse
|
122
|
Mutation-Driven Evolution of Pseudomonas aeruginosa in the Presence of either Ceftazidime or Ceftazidime-Avibactam. Antimicrob Agents Chemother 2018; 62:AAC.01379-18. [PMID: 30082283 DOI: 10.1128/aac.01379-18] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 07/27/2018] [Indexed: 02/04/2023] Open
Abstract
Ceftazidime-avibactam is a combination of β-lactam/β-lactamase inhibitor, the use of which is restricted to some clinical cases, including cystic fibrosis patients infected with multidrug-resistant Pseudomonas aeruginosa, in which mutation is the main driver of resistance. This study aims to predict the mechanisms of mutation-driven resistance that are selected for when P. aeruginosa is challenged with either ceftazidime or ceftazidime-avibactam. For this purpose, P. aeruginosa PA14 was submitted to experimental evolution in the absence of antibiotics and in the presence of increasing concentrations of ceftazidime or ceftazidime-avibactam for 30 consecutive days. Final populations were analyzed by whole-genome sequencing. All evolved populations reached similar levels of ceftazidime resistance. In addition, they were more susceptible to amikacin and produced pyomelanin. A first event in this evolution was the selection of large chromosomal deletions containing hmgA (involved in pyomelanin production), galU (involved in β-lactams resistance), and mexXY-oprM (involved in aminoglycoside resistance). Besides mutations in mpl and dacB that regulate β-lactamase expression, mutations related to MexAB-OprM overexpression were prevalent. Ceftazidime-avibactam challenge selected mutants in the putative efflux pump PA14_45890 and PA14_45910 and in a two-component system (PA14_45870 and PA14_45880), likely regulating its expression. All populations produced pyomelanin and were more susceptible to aminoglycosides, likely due to the selection of large chromosomal deletions. Since pyomelanin-producing mutants presenting similar deletions are regularly isolated from infections, the potential aminoglycoside hypersusceptiblity and reduced β-lactam susceptibility of pyomelanin-producing P. aeruginosa should be taken into consideration for treating infections caused by these isolates.
Collapse
|
123
|
Argáez-Correa W, Alvarez-Sánchez ME, Arana-Argáez VE, Ramírez-Camacho MA, Novelo-Castilla JS, Coral-Martínez TI, Torres-Romero JC. The Role of Iron Status in the Early Progression of Metronidazole Resistance in Trichomonas vaginalis Under Microaerophilic Conditions. J Eukaryot Microbiol 2018; 66:309-315. [PMID: 30047563 DOI: 10.1111/jeu.12671] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 06/14/2018] [Accepted: 07/18/2018] [Indexed: 01/22/2023]
Abstract
Trichomonas vaginalis is the etiological agent of human trichomoniasis. Metronidazole has high treatment success rate among trichomoniasis patients. However, metronidazole-resistant T. vaginalis has been reported, contributing in an increasing number of refractory cases. The mechanism of metronidazole resistance in this parasite is still unclear. In the vaginal environment, where the microaerophilic conditions prevail but the iron concentration is constantly fluctuating, the metronidazole resistance profile of T. vaginalis could be altered. In this study, we developed metronidazole-resistant strains of T. vaginalis and evaluate if iron availability is important to the action of the drug. The modulation of iron levels and iron chelation affected the actions of metronidazole both in susceptible and resistant strains. Interestingly, the early resistant strains exhibited minor iron content. The results of transcription analysis in the early resistant strains showed dysregulation in the expression of genes that codified proteins involved in iron transporter, iron-sulfur cluster assemblage, and oxidative stress response, which could not be observed in the late resistant and susceptible strains. Our results indicate that iron content plays an important role in the metronidazole action in T. vaginalis and likely to be related to iron-sulfur proteins involved in metronidazole activation and oxidative stress via Fenton reaction.
Collapse
Affiliation(s)
- Wendy Argáez-Correa
- Laboratorio de Bioquímica y Genética Molecular, Facultad de Química de la Universidad Autónoma de Yucatán, Mérida, 97069, Yucatán, México
| | - María E Alvarez-Sánchez
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, México City, 03100, México
| | - Victor E Arana-Argáez
- Laboratorio de Farmacología, Facultad de Química de la Universidad Autónoma de Yucatán, Mérida, 97069, Yucatán, México
| | - Mario A Ramírez-Camacho
- Centro de Información de Medicamentos, Facultad de Química de la Universidad Autónoma de Yucatán, Mérida, 97069, Yucatán, México
| | - Jazmín S Novelo-Castilla
- Laboratorio de Espectroscopía Atómica, Facultad de Química de la Universidad Autónoma de Yucatán, Mérida, 97069, Yucatán, México
| | - Tania I Coral-Martínez
- Laboratorio de Cromatografía, Facultad de Química de la Universidad Autónoma de Yucatán, Mérida, 97069, Yucatán, México
| | - Julio C Torres-Romero
- Laboratorio de Bioquímica y Genética Molecular, Facultad de Química de la Universidad Autónoma de Yucatán, Mérida, 97069, Yucatán, México
| |
Collapse
|
124
|
Kaplan E, Marano RBM, Jurkevitch E, Cytryn E. Enhanced Bacterial Fitness Under Residual Fluoroquinolone Concentrations Is Associated With Increased Gene Expression in Wastewater-Derived qnr Plasmid-Harboring Strains. Front Microbiol 2018; 9:1176. [PMID: 29937755 PMCID: PMC6003256 DOI: 10.3389/fmicb.2018.01176] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 05/15/2018] [Indexed: 12/18/2022] Open
Abstract
Plasmids harboring qnr genes confer resistance to low fluoroquinolone concentrations. These genes are of significant clinical, evolutionary and environmental importance, since they are widely distributed in a diverse array of natural and clinical environments. We previously extracted and sequenced a large (∼185 Kbp) qnrB-harboring plasmid, and several small (∼8 Kbp) qnrS-harboring plasmids, from Klebsiella pneumoniae isolates from municipal wastewater biosolids, and hypothesized that these plasmids provide host bacteria a selective advantage in wastewater treatment plants (WWTPs) that often contain residual concentrations of fluoroquinolones. The objectives of this study were therefore to determine the effect of residual fluoroquinolone concentrations on the growth kinetics of qnr plasmid-harboring bacteria; and on the copy number of qnr plasmids and expression of qnr genes. Electrotransformants harboring either one of the two types of plasmids could grow at ciprofloxacin concentrations exceeding 0.5 μg ml-1, but growth was significantly decreased at concentrations higher than 0.1 μg ml-1. In contrast, plasmid-free strains failed to grow even at 0.05 μg ml-1. No differences were observed in plasmid copy number under the tested ciprofloxacin concentrations, but qnr expression increased incrementally from 0 to 0.4 μg ml-1, suggesting that the transcription of this gene is regulated by antibiotic concentration. This study reveals that wastewater-derived qnr plasmids confer a selective advantage in the presence of residual fluoroquinolone concentrations and provides a mechanistic explanation for this phenomenon.
Collapse
Affiliation(s)
- Ella Kaplan
- Department of Soil Chemistry, Plant Nutrition and Microbiology, Institute of Soil, Water and Environmental Sciences, Volcani Center, Agricultural Research Organization, Beit Dagan, Israel.,Department of Agroecology and Plant Health, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Roberto B M Marano
- Department of Soil Chemistry, Plant Nutrition and Microbiology, Institute of Soil, Water and Environmental Sciences, Volcani Center, Agricultural Research Organization, Beit Dagan, Israel.,Department of Agroecology and Plant Health, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Edouard Jurkevitch
- Department of Soil Chemistry, Plant Nutrition and Microbiology, Institute of Soil, Water and Environmental Sciences, Volcani Center, Agricultural Research Organization, Beit Dagan, Israel
| | - Eddie Cytryn
- Department of Soil Chemistry, Plant Nutrition and Microbiology, Institute of Soil, Water and Environmental Sciences, Volcani Center, Agricultural Research Organization, Beit Dagan, Israel
| |
Collapse
|
125
|
Kiamco MM, Mohamed A, Reardon PN, Marean-Reardon CL, Aframehr WM, Call DR, Beyenal H, Renslow RS. Structural and metabolic responses of Staphylococcus aureus biofilms to hyperosmotic and antibiotic stress. Biotechnol Bioeng 2018; 115:1594-1603. [PMID: 29460278 PMCID: PMC5959008 DOI: 10.1002/bit.26572] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 01/10/2018] [Accepted: 02/08/2018] [Indexed: 01/26/2023]
Abstract
Biofilms alter their metabolism in response to environmental stress. This study explores the effect of a hyperosmotic agent-antibiotic treatment on the metabolism of Staphylococcus aureus biofilms through the use of nuclear magnetic resonance (NMR) techniques. To determine the metabolic activity of S. aureus, we quantified the concentrations of metabolites in spent medium using high-resolution NMR spectroscopy. Biofilm porosity, thickness, biovolume, and relative diffusion coefficient depth profiles were obtained using NMR microimaging. Dissolved oxygen concentration was measured to determine the availability of oxygen within the biofilm. Under vancomycin-only treatment, the biofilm communities switched to fermentation under anaerobic condition, as evidenced by high concentrations of formate (7.4 ± 2.7 mM), acetate (13.1 ± 0.9 mM), and lactate (3.0 ± 0.8 mM), and there was no detectable dissolved oxygen in the biofilm. In addition, we observed the highest consumption of pyruvate (0.19 mM remaining from an initial 40 mM concentration), the sole carbon source, under the vancomycin-only treatment. On the other hand, relative effective diffusion coefficients increased from 0.73 ± 0.08 to 0.88 ± 0.08 under vancomycin-only treatment but decreased from 0.71 ± 0.04 to 0.60 ± 0.07 under maltodextrin-only and from 0.73 ± 0.06 to 0.56 ± 0.08 under combined treatments. There was an increase in biovolume, from 2.5 ± 1 mm3 to 7 ± 1 mm3 , under the vancomycin-only treatment, while the maltodextrin-only and combined treatments showed no significant change in biovolume over time. This indicated that physical biofilm growth was halted during maltodextrin-only and combined treatments.
Collapse
Affiliation(s)
- Mia M Kiamco
- The Gene and Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, Washington
| | - Abdelrhman Mohamed
- The Gene and Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, Washington
| | - Patrick N Reardon
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington
| | - Carrie L Marean-Reardon
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington
| | - Wrya M Aframehr
- The Gene and Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, Washington
| | - Douglas R Call
- Paul G. Allen School for Global Animal Health, Washington State University, Pullman, Washington
| | - Haluk Beyenal
- The Gene and Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, Washington
| | - Ryan S Renslow
- The Gene and Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, Washington
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington
- Earth and Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington
| |
Collapse
|
126
|
Dunphy LJ, Papin JA. Biomedical applications of genome-scale metabolic network reconstructions of human pathogens. Curr Opin Biotechnol 2018; 51:70-79. [PMID: 29223465 PMCID: PMC5991985 DOI: 10.1016/j.copbio.2017.11.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 11/22/2017] [Accepted: 11/24/2017] [Indexed: 12/14/2022]
Abstract
The growing global threat of antibiotic resistant human pathogens has coincided with improved methods for developing and using genome-scale metabolic network reconstructions. Consequently, there has been an increase in the number of high-quality reconstructions of relevant human and zoonotic pathogens. Novel biomedical applications of pathogen reconstructions focus on three key aspects of pathogen behavior: the evolution of antibiotic resistance, virulence factor production, and host-pathogen interactions. New methods using these reconstructions aim to improve understanding of microbe pathogenicity and guide the development of new therapeutic strategies. This review summarizes the latest ways that genome-scale metabolic network reconstructions have been used to study human pathogens and suggests future applications with the potential to mitigate infectious disease.
Collapse
Affiliation(s)
- Laura J Dunphy
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22903, USA
| | - Jason A Papin
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22903, USA; Department of Medicine, Infectious Diseases and International Health, University of Virginia, Charlottesville, VA 22903, USA.
| |
Collapse
|
127
|
Yang K, Han Q, Chen B, Zheng Y, Zhang K, Li Q, Wang J. Antimicrobial hydrogels: promising materials for medical application. Int J Nanomedicine 2018; 13:2217-2263. [PMID: 29695904 PMCID: PMC5905846 DOI: 10.2147/ijn.s154748] [Citation(s) in RCA: 211] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The rapid emergence of antibiotic resistance in pathogenic microbes is becoming an imminent global public health problem. Local application of antibiotics might be a solution. In local application, materials need to act as the drug delivery system. The drug delivery system should be biodegradable and prolonged antibacterial effect should be provided to satisfy clinical demand. Hydrogel is a promising material for local antibacterial application. Hydrogel refers to a kind of biomaterial synthesized by a water-soluble natural polymer or a synthesized polymer, which turns into gel according to the change in different signals such as temperature, ionic strength, pH, ultraviolet exposure etc. Because of its high hydrophilicity, unique three-dimensional network, fine biocompatibility and cell adhesion, hydrogel is one of the suitable biomaterials for drug delivery in antimicrobial areas. In this review, studies from the past 5 years were reviewed, and several types of antimicrobial hydrogels according to different ingredients, different preparations, different antimicrobial mechanisms, different antimicrobial agents they contained and different applications, were summarized. The hydrogels loaded with metal nanoparticles as a potential method to solve antibiotic resistance were highlighted. Finally, future prospects of development and application of antimicrobial hydrogels are suggested.
Collapse
Affiliation(s)
- Kerong Yang
- Department of Orthopedics, The Second Hospital, Jilin University, Changchun, Jilin, People's Republic of China
| | - Qing Han
- Department of Orthopedics, The Second Hospital, Jilin University, Changchun, Jilin, People's Republic of China
| | - Bingpeng Chen
- Department of Orthopedics, The Second Hospital, Jilin University, Changchun, Jilin, People's Republic of China
| | - Yuhao Zheng
- Department of Orthopedics, The Second Hospital, Jilin University, Changchun, Jilin, People's Republic of China
| | - Kesong Zhang
- Department of Orthopedics, The Second Hospital, Jilin University, Changchun, Jilin, People's Republic of China
| | - Qiang Li
- Department of Orthopedics, The Second Hospital, Jilin University, Changchun, Jilin, People's Republic of China
| | - Jincheng Wang
- Department of Orthopedics, The Second Hospital, Jilin University, Changchun, Jilin, People's Republic of China
| |
Collapse
|
128
|
Shin B, Park C, Imlay JA, Park W. 4-Hydroxybenzaldehyde sensitizes Acinetobacter baumannii to amphenicols. Appl Microbiol Biotechnol 2018; 102:2323-2335. [PMID: 29387955 DOI: 10.1007/s00253-018-8791-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 01/02/2018] [Accepted: 01/16/2018] [Indexed: 12/21/2022]
Abstract
Bacterial metabolism modulated by environmental chemicals could alter antibiotic susceptibility. 4-Hydroxybenzaldehyde (4-HBA), which cannot support the growth of Acinetobacter baumannii, exhibited synergism only with amphenicol antibiotics including chloramphenicol (CAM) and thiamphenicol. Interestingly, this synergistic effect was not observed with other growth-supporting, structurally similar compounds such as 4-hydroxybenzoate. Transcriptomic analysis demonstrated that genes involved in protocatechuate metabolism (pca genes) and osmotic stress (bet genes) were significantly upregulated by 4-HBA and CAM treatment. The 14C-labeled CAM influx was lower in a pcaK1 (encoding a transporter of protocatechuate) deletion mutant and was higher in the pcaK1 overexpressing cells relative to that in the wild type upon 4-HBA treatment. Our kinetic data using 14C-labeled CAM clearly showed that CAM uptake is possibly through facilitated diffusion. Deletion of pcaK1 did not result in the elimination of CAM influx, indicating that CAM does not enter only through PcaK1. The amount of 4-HBA in the culture supernatant was, however, unaffected during the test conditions, validating that it was not metabolized by the bacteria. CAM resistant A. baumannii cells derived by serial passages through CAM-amended media exhibited lower level of pcaK1 gene expression. These results led us to conclude that the activation of PcaK1 transporter is probably linked to cellular CAM susceptibility. This is the first report showing a relationship between CAM influx and aromatic compound metabolism in A. baumannii.
Collapse
Affiliation(s)
- Bora Shin
- Laboratory of Molecular Environmental Microbiology, Department of Environmental Sciences and Ecological Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Chulwoo Park
- Laboratory of Molecular Environmental Microbiology, Department of Environmental Sciences and Ecological Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - James A Imlay
- Department of Microbiology, University of Illinois, Urbana, IL, 61801, USA
| | - Woojun Park
- Laboratory of Molecular Environmental Microbiology, Department of Environmental Sciences and Ecological Engineering, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
129
|
Correia S, Hébraud M, Poeta P, Capelo JL, Igrejas G. How combined multicomparative proteomic approaches can improve the understanding of quinolone resistance in Salmonella Typhimurium. Future Microbiol 2018; 13:403-406. [DOI: 10.2217/fmb-2017-0250] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Affiliation(s)
- Susana Correia
- Functional Genomics & Proteomics Unit, University of Trás-os-Montes & Alto Douro, Vila Real, Portugal
- Department of Genetics & Biotechnology, University of Trás-os-Montes & Alto Douro, Vila Real, Portugal
- Veterinary Science Department, University of Trás-os-Montes & Alto Douro, Vila Real, Portugal
- LAQV-REQUIMTE, Faculty of Science & Technology, Nova University of Lisbon, Caparica, Portugal
| | - Michel Hébraud
- UMR454 MEDiS, Institut National de la Recherche Agronomique (INRA), Centre Auvergne-Rhône-Alpes, site de Theix, Saint-Genès Champanelle, France
- Plate-Forme d’Exploration du Métabolisme composante protéomique, UR370 QuaPA, Institut National de la Recherche Agronomique (INRA), Centre Auvergne-Rhône-Alpes, site de Theix, Saint-Genès Champanelle, France
| | - Patrícia Poeta
- Veterinary Science Department, University of Trás-os-Montes & Alto Douro, Vila Real, Portugal
- LAQV-REQUIMTE, Faculty of Science & Technology, Nova University of Lisbon, Caparica, Portugal
| | - José L Capelo
- LAQV-REQUIMTE, Faculty of Science & Technology, Nova University of Lisbon, Caparica, Portugal
- ProteoMass Scientific Society, Faculty of Sciences & Technology, Campus de Caparica, Caparica, Portugal
| | - Gilberto Igrejas
- Functional Genomics & Proteomics Unit, University of Trás-os-Montes & Alto Douro, Vila Real, Portugal
- Department of Genetics & Biotechnology, University of Trás-os-Montes & Alto Douro, Vila Real, Portugal
- LAQV-REQUIMTE, Faculty of Science & Technology, Nova University of Lisbon, Caparica, Portugal
| |
Collapse
|
130
|
Yamamoto K, Yamanaka Y, Shimada T, Sarkar P, Yoshida M, Bhardwaj N, Watanabe H, Taira Y, Chatterji D, Ishihama A. Altered Distribution of RNA Polymerase Lacking the Omega Subunit within the Prophages along the Escherichia coli K-12 Genome. mSystems 2018; 3:e00172-17. [PMID: 29468196 PMCID: PMC5811629 DOI: 10.1128/msystems.00172-17] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 01/25/2018] [Indexed: 11/20/2022] Open
Abstract
The RNA polymerase (RNAP) of Escherichia coli K-12 is a complex enzyme consisting of the core enzyme with the subunit structure α2ββ'ω and one of the σ subunits with promoter recognition properties. The smallest subunit, omega (the rpoZ gene product), participates in subunit assembly by supporting the folding of the largest subunit, β', but its functional role remains unsolved except for its involvement in ppGpp binding and stringent response. As an initial approach for elucidation of its functional role, we performed in this study ChIP-chip (chromatin immunoprecipitation with microarray technology) analysis of wild-type and rpoZ-defective mutant strains. The altered distribution of RpoZ-defective RNAP was identified mostly within open reading frames, in particular, of the genes inside prophages. For the genes that exhibited increased or decreased distribution of RpoZ-defective RNAP, the level of transcripts increased or decreased, respectively, as detected by reverse transcription-quantitative PCR (qRT-PCR). In parallel, we analyzed, using genomic SELEX (systemic evolution of ligands by exponential enrichment), the distribution of constitutive promoters that are recognized by RNAP RpoD holoenzyme alone and of general silencer H-NS within prophages. Since all 10 prophages in E. coli K-12 carry only a small number of promoters, the altered occupancy of RpoZ-defective RNAP and of transcripts might represent transcription initiated from as-yet-unidentified host promoters. The genes that exhibited transcription enhanced by RpoZ-defective RNAP are located in the regions of low-level H-NS binding. By using phenotype microarray (PM) assay, alterations of some phenotypes were detected for the rpoZ-deleted mutant, indicating the involvement of RpoZ in regulation of some genes. Possible mechanisms of altered distribution of RNAP inside prophages are discussed. IMPORTANCE The 91-amino-acid-residue small-subunit omega (the rpoZ gene product) of Escherichia coli RNA polymerase plays a structural role in the formation of RNA polymerase (RNAP) as a chaperone in folding the largest subunit (β', of 1,407 residues in length), but except for binding of the stringent signal ppGpp, little is known of its role in the control of RNAP function. After analysis of genomewide distribution of wild-type and RpoZ-defective RNAP by the ChIP-chip method, we found alteration of the RpoZ-defective RNAP inside open reading frames, in particular, of the genes within prophages. For a set of the genes that exhibited altered occupancy of the RpoZ-defective RNAP, transcription was found to be altered as observed by qRT-PCR assay. All the observations here described indicate the involvement of RpoZ in recognition of some of the prophage genes. This study advances understanding of not only the regulatory role of omega subunit in the functions of RNAP but also the regulatory interplay between prophages and the host E. coli for adjustment of cellular physiology to a variety of environments in nature.
Collapse
Affiliation(s)
- Kaneyoshi Yamamoto
- Department of Frontier Bioscience, Hosei University, Tokyo, Japan
- Micro-Nano Technology Research Center, Hosei University, Tokyo, Japan
| | - Yuki Yamanaka
- Micro-Nano Technology Research Center, Hosei University, Tokyo, Japan
| | - Tomohiro Shimada
- Micro-Nano Technology Research Center, Hosei University, Tokyo, Japan
- Meiji University, School of Agriculture, Kawasaki, Kanagawa, Japan
| | - Paramita Sarkar
- Department of Frontier Bioscience, Hosei University, Tokyo, Japan
- Indian Institute of Science, Molecular Biophysics Unit, Bangalore, India
| | - Myu Yoshida
- Department of Frontier Bioscience, Hosei University, Tokyo, Japan
| | - Neerupma Bhardwaj
- Indian Institute of Science, Molecular Biophysics Unit, Bangalore, India
| | - Hiroki Watanabe
- Department of Frontier Bioscience, Hosei University, Tokyo, Japan
| | - Yuki Taira
- Department of Frontier Bioscience, Hosei University, Tokyo, Japan
| | - Dipankar Chatterji
- Indian Institute of Science, Molecular Biophysics Unit, Bangalore, India
| | - Akira Ishihama
- Micro-Nano Technology Research Center, Hosei University, Tokyo, Japan
| |
Collapse
|
131
|
Abstract
Antibiotics act on bacterial metabolism, and antibiotic resistance involves changes in this metabolism. Interventions on metabolism with drugs might therefore modify drug susceptibility and drug resistance. In their recent article, Martin Vestergaard et al. (mBio 8:e01114-17, 2017, https://doi.org/10.1128/mBio.01114-17) illustrate the possibility of converting intrinsically resistant bacteria into susceptible ones. They reported that inhibition of a central metabolic enzyme, ATP synthase, allows otherwise ineffective polymyxin antibiotics to act on Staphylococcus aureus The study of the intrinsic resistome of bacterial pathogens has shown that several metabolic genes, including multigene transcriptional regulators, contribute to antibiotic resistance. In some cases, these genes only marginally increase antibiotic resistance, but reduced levels of susceptibility might be critical in the evolution or resistance under low antibiotic concentrations or in the clinical response of highly resistant bacteria. Drug interventions on bacterial metabolism might constitute a critical adjuvant therapy in combination with antibiotics to ensure susceptibility of pathogens with intrinsic or acquired antimicrobial resistance.
Collapse
|
132
|
Cardona ST, Choy M, Hogan AM. Essential Two-Component Systems Regulating Cell Envelope Functions: Opportunities for Novel Antibiotic Therapies. J Membr Biol 2017; 251:75-89. [DOI: 10.1007/s00232-017-9995-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 10/20/2017] [Indexed: 01/22/2023]
|
133
|
Correia S, Hébraud M, Chafsey I, Chambon C, Viala D, Torres C, Caniça M, Capelo JL, Poeta P, Igrejas G. Subproteomic signature comparison of in vitro selected fluoroquinolone resistance and ciprofloxacin stress in Salmonella Typhimurium DT104B. Expert Rev Proteomics 2017; 14:941-961. [PMID: 28871888 DOI: 10.1080/14789450.2017.1375856] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Fluoroquinolone resistance in nontyphoidal Salmonella is a situation of serious and international concern, particularly in S. Typhimurium DT104B multiresistant strains. Although known to be multifactorial, fluoroquinolone resistance is still far from a complete understanding. METHODS Subproteome changes between an experimentally selected fluoroquinolone-resistant strain (Se6-M) and its parent strain (Se6), and also in Se6-M under ciprofloxacin (CIP) stress, were evaluated in order to give new insights into the mechanisms involved. Proteomes were compared at the intracellular and membrane levels by a 2-DE~LC-MS/MS and a shotgun LC-MS/MS approach, respectively. RESULTS In total, 35 differentially abundant proteins were identified when comparing Se6 with Se6-M (25 more abundant in Se6 and 10 more abundant in Se6-M) and 82 were identified between Se6-M and Se6-M+CIP (51 more abundant in Se6-M and 31 more abundant under ciprofloxacin stress). CONCLUSION Several proteins with known and possible roles in quinolone resistance were identified which provide important information about mechanism-related differential protein expression, supporting the current knowledge and also leading to new testable hypotheses on the mechanism of action of fluoroquinolone drugs.
Collapse
Affiliation(s)
- Susana Correia
- a Functional Genomics and Proteomics Unit , University of Trás-os-Montes and Alto Douro , Vila Real , Portugal.,b Department of Genetics and Biotechnology , University of Trás-os-Montes and Alto Douro , Vila Real , Portugal.,c Veterinary Science Department , University of Trás-os-Montes and Alto Douro , Vila Real , Portugal.,d UCIBIO-REQUIMTE, Faculty of Science and Technology , Nova University of Lisbon , Caparica , Portugal
| | - Michel Hébraud
- e UR454 Microbiology , Institut National de la Recherche Agronomique (INRA), Centre Auvergne-Rhône-Alpes , site de Theix , Saint-Genès Champanelle , France.,f Plate-Forme d'Exploration du Métabolisme composante protéomique, UR370 QuaPA , Institut National de la Recherche Agronomique (INRA), Centre Auvergne-Rhône-Alpes , site de Theix , Saint-Genès Champanelle , France
| | - Ingrid Chafsey
- e UR454 Microbiology , Institut National de la Recherche Agronomique (INRA), Centre Auvergne-Rhône-Alpes , site de Theix , Saint-Genès Champanelle , France
| | - Christophe Chambon
- f Plate-Forme d'Exploration du Métabolisme composante protéomique, UR370 QuaPA , Institut National de la Recherche Agronomique (INRA), Centre Auvergne-Rhône-Alpes , site de Theix , Saint-Genès Champanelle , France
| | - Didier Viala
- f Plate-Forme d'Exploration du Métabolisme composante protéomique, UR370 QuaPA , Institut National de la Recherche Agronomique (INRA), Centre Auvergne-Rhône-Alpes , site de Theix , Saint-Genès Champanelle , France
| | - Carmen Torres
- g Área de Bioquímica y Biología Molecular , Universidad de La Rioja , Logroño , Spain
| | - Manuela Caniça
- h National Reference Laboratory of Antibiotic Resistances and Healthcare Associated Infections (NRL-AMR-HAI), Department of Infectious Diseases , National Health Institute Doutor Ricardo Jorge (INSA) , Lisbon , Portugal
| | - José Luis Capelo
- d UCIBIO-REQUIMTE, Faculty of Science and Technology , Nova University of Lisbon , Caparica , Portugal.,i Faculty of Sciences and Technology , ProteoMass Scientific Society , Caparica , Portugal
| | - Patrícia Poeta
- c Veterinary Science Department , University of Trás-os-Montes and Alto Douro , Vila Real , Portugal.,d UCIBIO-REQUIMTE, Faculty of Science and Technology , Nova University of Lisbon , Caparica , Portugal
| | - Gilberto Igrejas
- a Functional Genomics and Proteomics Unit , University of Trás-os-Montes and Alto Douro , Vila Real , Portugal.,b Department of Genetics and Biotechnology , University of Trás-os-Montes and Alto Douro , Vila Real , Portugal.,d UCIBIO-REQUIMTE, Faculty of Science and Technology , Nova University of Lisbon , Caparica , Portugal
| |
Collapse
|
134
|
Li J, Hao H, Cheng G, Liu C, Ahmed S, Shabbir MAB, Hussain HI, Dai M, Yuan Z. Microbial Shifts in the Intestinal Microbiota of Salmonella Infected Chickens in Response to Enrofloxacin. Front Microbiol 2017; 8:1711. [PMID: 28943868 PMCID: PMC5596078 DOI: 10.3389/fmicb.2017.01711] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Accepted: 08/24/2017] [Indexed: 12/18/2022] Open
Abstract
Fluoroquinolones (FQs) are important antibiotics used for treatment of Salmonella infection in poultry in many countries. However, oral administration of fluoroquinolones may affect the composition and abundance of a number of bacterial taxa in the chicken intestine. Using 16S rRNA gene sequencing, the microbial shifts in the gut of Salmonella infected chickens in response to enrofloxacin treatments at different dosages (0, 0.1, 4, and 100 mg/kg b.w.) were quantitatively evaluated. The results showed that the shedding levels of Salmonella were significantly reduced in the high dosage group as demonstrated by both the culturing method and 16S rRNA sequencing method. The average values of diversity indices were higher in the control group than in the three medicated groups. Non-metric multidimensional scaling (NMDS) analysis results showed that the microbial community of high dosage group was clearly separated from the other three groups. In total, 25 genera were significantly enriched (including 6 abundant genera: Lactococcus, Bacillus, Burkholderia, Pseudomonas, Rhizobium, and Acinetobacter) and 23 genera were significantly reduced in the medicated groups than in the control group for the treatment period, but these bacterial taxa recovered to normal levels after therapy withdrawal. Additionally, 5 genera were significantly reduced in both treatment and withdrawal periods (e.g., Blautia and Anaerotruncus) and 23 genera (e.g., Enterobacter and Clostridium) were significantly decreased only in the withdrawal period, indicating that these genera might be the potential targets for the fluoroquinolones antimicrobial effects. Specially, Enterococcus was significantly reduced under high dosage of enrofloxacin treatment, while significantly enriched in the withdrawal period, which was presumably due to the resistance selection. Predicted microbial functions associated with genetic information processing were significantly decreased in the high dosage group. Overall, enrofloxacin at a dosage of 100 mg/kg b.w. significantly altered the microbial community membership and structure, and microbial functions in the chicken intestine during the medication. This study fully investigates the chicken intestinal microbiota in response to enrofloxacin treatment and identifies potential targets against which the fluoroquinolones may have potent antimicrobial effects. These results provide insights into the effects of the usage of enrofloxacin on chicken and will aid in the prudent and rational use of antibiotics in poultry industry.
Collapse
Affiliation(s)
- Jun Li
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MOA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural UniversityWuhan, China
| | - Haihong Hao
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural UniversityWuhan, China
| | - Guyue Cheng
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural UniversityWuhan, China
| | - Chunbei Liu
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MOA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural UniversityWuhan, China
| | - Saeed Ahmed
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MOA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural UniversityWuhan, China
| | - Muhammad A B Shabbir
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MOA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural UniversityWuhan, China
| | - Hafiz I Hussain
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MOA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural UniversityWuhan, China
| | - Menghong Dai
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural UniversityWuhan, China
| | - Zonghui Yuan
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MOA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural UniversityWuhan, China.,MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural UniversityWuhan, China
| |
Collapse
|
135
|
Irnov I, Wang Z, Jannetty ND, Bustamante JA, Rhee KY, Jacobs-Wagner C. Crosstalk between the tricarboxylic acid cycle and peptidoglycan synthesis in Caulobacter crescentus through the homeostatic control of α-ketoglutarate. PLoS Genet 2017; 13:e1006978. [PMID: 28827812 PMCID: PMC5578688 DOI: 10.1371/journal.pgen.1006978] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 08/31/2017] [Accepted: 08/15/2017] [Indexed: 11/18/2022] Open
Abstract
To achieve robust replication, bacteria must integrate cellular metabolism and cell wall growth. While these two processes have been well characterized, the nature and extent of cross-regulation between them is not well understood. Here, using classical genetics, CRISPRi, metabolomics, transcriptomics and chemical complementation approaches, we show that a loss of the master regulator Hfq in Caulobacter crescentus alters central metabolism and results in cell shape defects in a nutrient-dependent manner. We demonstrate that the cell morphology phenotype in the hfq deletion mutant is attributable to a disruption of α-ketoglutarate (KG) homeostasis. In addition to serving as a key intermediate of the tricarboxylic acid (TCA) cycle, KG is a by-product of an enzymatic reaction required for the synthesis of peptidoglycan, a major component of the bacterial cell wall. Accumulation of KG in the hfq deletion mutant interferes with peptidoglycan synthesis, resulting in cell morphology defects and increased susceptibility to peptidoglycan-targeting antibiotics. This work thus reveals a direct crosstalk between the TCA cycle and cell wall morphogenesis. This crosstalk highlights the importance of metabolic homeostasis in not only ensuring adequate availability of biosynthetic precursors, but also in preventing interference with cellular processes in which these intermediates arise as by-products.
Collapse
Affiliation(s)
- Irnov Irnov
- Microbial Sciences Institute, Yale University, West Haven, CT, United States of America
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, United States of America
| | - Zhe Wang
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY, United States of America
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medical College, New York, NY, United States of America
| | - Nicholas D. Jannetty
- Microbial Sciences Institute, Yale University, West Haven, CT, United States of America
- Howard Hughes Medical Institute, Yale University, New Haven, CT, United States of America
| | - Julian A. Bustamante
- Howard Hughes Medical Institute, Yale University, New Haven, CT, United States of America
| | - Kyu Y. Rhee
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY, United States of America
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medical College, New York, NY, United States of America
| | - Christine Jacobs-Wagner
- Microbial Sciences Institute, Yale University, West Haven, CT, United States of America
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, United States of America
- Howard Hughes Medical Institute, Yale University, New Haven, CT, United States of America
- Department of Microbial Pathogenesis, Yale School of Medicine, Yale University, New Haven, CT, United States of America
- * E-mail:
| |
Collapse
|
136
|
King JR, Woolston BM, Stephanopoulos G. Designing a New Entry Point into Isoprenoid Metabolism by Exploiting Fructose-6-Phosphate Aldolase Side Reactivity of Escherichia coli. ACS Synth Biol 2017; 6:1416-1426. [PMID: 28375628 DOI: 10.1021/acssynbio.7b00072] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The 2C-methyl-d-erythritol-4-phosphate (MEP) pathway in Escherichia coli has been highlighted for its potential to provide access to myriad isoprenoid chemicals of industrial and therapeutic relevance and discover antibiotic targets to treat microbial human pathogens. Here, we describe a metabolic engineering strategy for the de novo construction of a biosynthetic pathway that produces 1-dexoxy-d-xylulose-5-phosphate (DXP), the precursor metabolite of the MEP pathway, from the simple and renewable starting materials d-arabinose and hydroxyacetone. Unlike most metabolic engineering efforts in which cell metabolism is reprogrammed with enzymes that are highly specific to their desired reaction, we highlight the promiscuous activity of the native E. coli fructose-6-phosphate aldolase as central to the metabolic rerouting of carbon to DXP. We use mass spectrometric isotopomer analysis of intracellular metabolites to show that the engineered pathway is able to support in vivo DXP biosynthesis in E. coli. The engineered DXP synthesis is further able to rescue cells that were chemically inhibited in their ability to produce DXP and to increase terpene titers in strains harboring the non-native lycopene pathway. In addition to providing an alternative metabolic pathway to produce isoprenoids, the results here highlight the potential role of pathway evolution to circumvent metabolic inhibitors in the development of microbial antibiotic resistance.
Collapse
Affiliation(s)
- Jason R. King
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
| | - Benjamin M. Woolston
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
| | - Gregory Stephanopoulos
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
137
|
Labella JI, Cantos R, Espinosa J, Forcada-Nadal A, Rubio V, Contreras A. PipY, a Member of the Conserved COG0325 Family of PLP-Binding Proteins, Expands the Cyanobacterial Nitrogen Regulatory Network. Front Microbiol 2017; 8:1244. [PMID: 28744260 PMCID: PMC5504682 DOI: 10.3389/fmicb.2017.01244] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 06/20/2017] [Indexed: 11/13/2022] Open
Abstract
Synechococcus elongatus PCC 7942 is a paradigmatic model organism for nitrogen regulation in cyanobacteria. Expression of genes involved in nitrogen assimilation is positively regulated by the 2-oxoglutarate receptor and global transcriptional regulator NtcA. Maximal activation requires the subsequent binding of the co-activator PipX. PII, a protein found in all three domains of life as an integrator of signals of the nitrogen and carbon balance, binds to PipX to counteract NtcA activity at low 2-oxoglutarate levels. PII-PipX complexes can also bind to the transcriptional regulator PlmA, whose regulon remains unknown. Here we expand the nitrogen regulatory network to PipY, encoded by the bicistronic operon pipXY in S. elongatus. Work with PipY, the cyanobacterial member of the widespread family of COG0325 proteins, confirms the conserved roles in vitamin B6 and amino/keto acid homeostasis and reveals new PLP-related phenotypes, including sensitivity to antibiotics targeting essential PLP-holoenzymes or synthetic lethality with cysK. In addition, the related phenotypes of pipY and pipX mutants are consistent with genetic interactions in the contexts of survival to PLP-targeting antibiotics and transcriptional regulation. We also showed that PipY overexpression increased the length of S. elongatus cells. Taken together, our results support a universal regulatory role for COG0325 proteins, paving the way to a better understanding of these proteins and of their connections with other biological processes.
Collapse
Affiliation(s)
- José I Labella
- Departamento de Fisiología, Genética y Microbiología, Universidad de AlicanteAlicante, Spain
| | - Raquel Cantos
- Departamento de Fisiología, Genética y Microbiología, Universidad de AlicanteAlicante, Spain
| | - Javier Espinosa
- Departamento de Fisiología, Genética y Microbiología, Universidad de AlicanteAlicante, Spain
| | - Alicia Forcada-Nadal
- Departamento de Fisiología, Genética y Microbiología, Universidad de AlicanteAlicante, Spain.,Instituto de Biomedicina de Valencia - Consejo Superior de Investigaciones CientíficasValencia, Spain
| | - Vicente Rubio
- Instituto de Biomedicina de Valencia - Consejo Superior de Investigaciones CientíficasValencia, Spain.,Group 739, Centro de Investigación Biomédica en Red de Enfermedades Raras - Instituto de Salud Carlos IIIValencia, Spain
| | - Asunción Contreras
- Departamento de Fisiología, Genética y Microbiología, Universidad de AlicanteAlicante, Spain
| |
Collapse
|
138
|
Characterization of Thermophilic Bacteria Isolated from two Hot Springs in Jazan, Saudi Arabia. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2017. [DOI: 10.22207/jpam.11.2.13] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
139
|
Correia S, Hébraud M, Chafsey I, Chambon C, Viala D, Sáenz Y, Capelo JL, Poeta P, Igrejas G. Comparative subproteomic analysis of clinically acquired fluoroquinolone resistance and ciprofloxacin stress in Salmonella Typhimurium DT104B. Proteomics Clin Appl 2017; 11. [PMID: 28314077 DOI: 10.1002/prca.201600107] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 02/21/2017] [Accepted: 03/07/2017] [Indexed: 02/06/2023]
Abstract
PURPOSE Antimicrobial resistance is a worldwide public health threat and Salmonella enterica subsp. enterica serotype Typhimurium phage type DT104B multiresistant strains with additional quinolone resistance have been responsible for global outbreaks and high mortality. Quinolone resistance is known to be multifactorial but is still far from a complete understanding. To give new insights about the resistance mechanisms involved, this work aimed to evaluate subproteome changes between an S. Typhimurium DT104B clinical strain that acquired fluoroquinolone resistance after treatment (Se20) and its pretreatment parental strain (Se6), and also subproteome variations in Se20 under ciprofloxacin (CIP) stress (Se20+CIP). EXPERIMENTAL DESIGN The proteomes were compared at the intracellular and membrane levels by a 2-DE∼LC-MS/MS and a shotgun LC-MS/MS approach, respectively. RESULTS In total, 14 differentially abundant proteins were identified when comparing Se6 with Se20 and 91 were identified between Se20 and Se20+CIP. Several proteins with known and possible roles in quinolone resistance (AAC(6')-Ib-cr4, OmpD, OmpX, GlmS, GlmU, H-NS, etc.) were identified and discussed. CONCLUSIONS AND CLINICAL RELEVANCE The great number of proteins identified in this study provides important information about mechanism-related differential protein expression which supports the current knowledge and might lead to new testable hypotheses on the mechanism of action of fluoroquinolone drugs.
Collapse
Affiliation(s)
- Susana Correia
- Functional Genomics and Proteomics Unit, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.,Department of Genetics and Biotechnology, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.,Veterinary Science Department, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.,UCIBIO-REQUIMTE, Faculty of Science and Technology, Nova University of Lisbon, Lisbon, Portugal
| | - Michel Hébraud
- Institut National de la Recherche Agronomique (INRA), Université Clermont Auvergne (UCA), UMR Microbiologie Environnement Digestif et Santé (MEDiS), site de Theix, France.,Institut National de la Recherche Agronomique (INRA), UR370 QuaPA, Plate-Forme d'Exploration du Métabolisme composante protéomique, site de Theix, France
| | - Ingrid Chafsey
- Institut National de la Recherche Agronomique (INRA), Université Clermont Auvergne (UCA), UMR Microbiologie Environnement Digestif et Santé (MEDiS), site de Theix, France
| | - Christophe Chambon
- Institut National de la Recherche Agronomique (INRA), UR370 QuaPA, Plate-Forme d'Exploration du Métabolisme composante protéomique, site de Theix, France
| | - Didier Viala
- Institut National de la Recherche Agronomique (INRA), UR370 QuaPA, Plate-Forme d'Exploration du Métabolisme composante protéomique, site de Theix, France
| | - Yolanda Sáenz
- Área de Microbiología Molecular, Centro de Investigación Biomédica de La Rioja, Logrono, Spain
| | - José Luis Capelo
- UCIBIO-REQUIMTE, Faculty of Science and Technology, Nova University of Lisbon, Lisbon, Portugal.,ProteoMass Scientific Society, Faculty of Sciences and Technology, Caparica Campus, Caparica, Portugal
| | - Patrícia Poeta
- Veterinary Science Department, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.,UCIBIO-REQUIMTE, Faculty of Science and Technology, Nova University of Lisbon, Lisbon, Portugal
| | - Gilberto Igrejas
- Functional Genomics and Proteomics Unit, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.,Department of Genetics and Biotechnology, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.,UCIBIO-REQUIMTE, Faculty of Science and Technology, Nova University of Lisbon, Lisbon, Portugal
| |
Collapse
|
140
|
Correia S, Poeta P, Hébraud M, Capelo JL, Igrejas G. Mechanisms of quinolone action and resistance: where do we stand? J Med Microbiol 2017; 66:551-559. [DOI: 10.1099/jmm.0.000475] [Citation(s) in RCA: 156] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Susana Correia
- Functional Genomics and Proteomics Unit, University of Trás-os-Montes and Alto Douro, Portugal
- Department of Genetics and Biotechnology, University of Trás-os-Montes and Alto Douro, Portugal
- UCIBIO-REQUIMTE, Faculty of Science and Technology, NOVA University of Lisbon, Lisbon, Portugal
- Veterinary Science Department, University of Trás-os-Montes and Alto Douro, Portugal
| | - Patrícia Poeta
- Veterinary Science Department, University of Trás-os-Montes and Alto Douro, Portugal
- UCIBIO-REQUIMTE, Faculty of Science and Technology, NOVA University of Lisbon, Lisbon, Portugal
| | - Michel Hébraud
- Université Clermont Auvergne (UCA), UMR Microbiologie Environnement Digestif et Santé (MEDiS), Institut National de la Recherche Agronomique (INRA), Centre Auvergne-Rhône-Alpes, site de Theix, France
- Plate-Forme d’Exploration du Métabolisme composante protéomique, UR370 QuaPA, Institut National de la Recherche Agronomique (INRA), Centre Auvergne-Rhône-Alpes, site de Theix, France
| | - José Luis Capelo
- UCIBIO-REQUIMTE, Faculty of Science and Technology, NOVA University of Lisbon, Lisbon, Portugal
- ProteoMass Scientific Society, Faculty of Sciences and Technology, Caparica Campus, Portugal
| | - Gilberto Igrejas
- Department of Genetics and Biotechnology, University of Trás-os-Montes and Alto Douro, Portugal
- Functional Genomics and Proteomics Unit, University of Trás-os-Montes and Alto Douro, Portugal
- UCIBIO-REQUIMTE, Faculty of Science and Technology, NOVA University of Lisbon, Lisbon, Portugal
| |
Collapse
|
141
|
Banerjee D, Parmar D, Bhattacharya N, Ghanate AD, Panchagnula V, Raghunathan A. A scalable metabolite supplementation strategy against antibiotic resistant pathogen Chromobacterium violaceum induced by NAD +/NADH + imbalance. BMC SYSTEMS BIOLOGY 2017; 11:51. [PMID: 28446174 PMCID: PMC5405553 DOI: 10.1186/s12918-017-0427-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 04/21/2017] [Indexed: 12/18/2022]
Abstract
BACKGROUND The leading edge of the global problem of antibiotic resistance necessitates novel therapeutic strategies. This study develops a novel systems biology driven approach for killing antibiotic resistant pathogens using benign metabolites. RESULTS Controlled laboratory evolutions established chloramphenicol and streptomycin resistant pathogens of Chromobacterium. These resistant pathogens showed higher growth rates and required higher lethal doses of antibiotic. Growth and viability testing identified malate, maleate, succinate, pyruvate and oxoadipate as resensitising agents for antibiotic therapy. Resistant genes were catalogued through whole genome sequencing. Intracellular metabolomic profiling identified violacein as a potential biomarker for resistance. The temporal variance of metabolites captured the linearized dynamics around the steady state and correlated to growth rate. A constraints-based flux balance model of the core metabolism was used to predict the metabolic basis of antibiotic susceptibility and resistance. CONCLUSIONS The model predicts electron imbalance and skewed NAD/NADH ratios as a result of antibiotics - chloramphenicol and streptomycin. The resistant pathogen rewired its metabolic networks to compensate for disruption of redox homeostasis. We foresee the utility of such scalable workflows in identifying metabolites for clinical isolates as inevitable solutions to mitigate antibiotic resistance.
Collapse
Affiliation(s)
- Deepanwita Banerjee
- Chemical Engineering Division, CSIR-National Chemical Laboratory, Pune, India
| | | | | | - Avinash D. Ghanate
- Chemical Engineering Division, CSIR-National Chemical Laboratory, Pune, India
| | | | - Anu Raghunathan
- Chemical Engineering Division, CSIR-National Chemical Laboratory, Pune, India
| |
Collapse
|
142
|
The Bifunctional Enzyme SpoT Is Involved in the Clarithromycin Tolerance of Helicobacter pylori by Upregulating the Transporters HP0939, HP1017, HP0497, and HP0471. Antimicrob Agents Chemother 2017; 61:AAC.02011-16. [PMID: 28242673 PMCID: PMC5404559 DOI: 10.1128/aac.02011-16] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 02/21/2017] [Indexed: 01/30/2023] Open
Abstract
Clarithromycin (CLA) is a commonly recommended drug for Helicobacter pylori eradication. However, the prevalence of CLA-resistant H. pylori is increasing. Although point mutations in the 23S rRNA are key factors for CLA resistance, other factors, including efflux pumps and regulation genes, are also involved in the resistance of H. pylori to CLA. Guanosine 3′-diphosphate 5′-triphosphate and guanosine 3′,5′-bispyrophosphate [(p)ppGpp)], which are synthesized by the bifunctional enzyme SpoT in H. pylori, play an important role for some bacteria to adapt to antibiotic pressure. Nevertheless, no related research involving H. pylori has been reported. In addition, transporters have been found to be related to bacterial drug resistance. Therefore, this study investigated the function of SpoT in H. pylori resistance to CLA by examining the shifts in the expression of transporters and explored the role of transporters in the CLA resistance of H. pylori. A ΔspoT strain was constructed in this study, and it was shown that SpoT is involved in H. pylori tolerance of CLA by upregulating the transporters HP0939, HP1017, HP0497, and HP0471. This was assessed using a series of molecular and biochemical experiments and a cDNA microarray. Additionally, the knockout of genes hp0939, hp0471, and hp0497 in the resistant strains caused a reduction or loss (the latter in the Δhp0497 strain) of resistance to CLA. Furthermore, the average expression levels of these four transporters in clinical CLA-resistant strains were considerably higher than those in clinical CLA-sensitive strains. Taken together, our results revealed a novel molecular mechanism of H. pylori adaption to CLA stress.
Collapse
|
143
|
Antibiotic stress selects against cooperation in the pathogenic bacterium Pseudomonas aeruginosa. Proc Natl Acad Sci U S A 2017; 114:546-551. [PMID: 28049833 DOI: 10.1073/pnas.1612522114] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Cheats are a pervasive threat to public goods production in natural and human communities, as they benefit from the commons without contributing to it. Although ecological antagonisms such as predation, parasitism, competition, and abiotic environmental stress play key roles in shaping population biology, it is unknown how such stresses generally affect the ability of cheats to undermine cooperation. We used theory and experiments to address this question in the pathogenic bacterium, Pseudomonas aeruginosa Although public goods producers were selected against in all populations, our competition experiments showed that antibiotics significantly increased the advantage of nonproducers. Moreover, the dominance of nonproducers in mixed cultures was associated with higher resistance to antibiotics than in either monoculture. Mathematical modeling indicates that accentuated costs to producer phenotypes underlie the observed patterns. Mathematical analysis further shows how these patterns should generalize to other taxa with public goods behaviors. Our findings suggest that explaining the maintenance of cooperative public goods behaviors in certain natural systems will be more challenging than previously thought. Our results also have specific implications for the control of pathogenic bacteria using antibiotics and for understanding natural bacterial ecosystems, where subinhibitory concentrations of antimicrobials frequently occur.
Collapse
|
144
|
Harrison JP, Angel R, Cockell CS. Astrobiology as a framework for investigating antibiotic susceptibility: a study of Halomonas hydrothermalis. J R Soc Interface 2017; 14:20160942. [PMID: 28123098 PMCID: PMC5310740 DOI: 10.1098/rsif.2016.0942] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 12/14/2016] [Indexed: 01/19/2023] Open
Abstract
Physical and chemical boundaries for microbial multiplication on Earth are strongly influenced by interactions between environmental extremes. However, little is known about how interactions between multiple stress parameters affect the sensitivity of microorganisms to antibiotics. Here, we assessed how 12 distinct permutations of salinity, availability of an essential nutrient (iron) and atmospheric composition (aerobic or microaerobic) affect the susceptibility of a polyextremotolerant bacterium, Halomonas hydrothermalis, to ampicillin, kanamycin and ofloxacin. While salinity had a significant impact on sensitivity to all three antibiotics (as shown by turbidimetric analyses), the nature of this impact was modified by iron availability and the ambient gas composition, with differing effects observed for each compound. These two parameters were found to be of particular importance when considered in combination and, in the case of ampicillin, had a stronger combined influence on antibiotic tolerance than salinity. Our data show how investigating microbial responses to multiple extremes, which are more representative of natural habitats than single extremes, can improve our understanding of the effects of antimicrobial compounds and suggest how studies of habitability, motivated by the desire to map the limits of life, can be used to systematically assess the effectiveness of antibiotics.
Collapse
Affiliation(s)
- Jesse P Harrison
- UK Centre for Astrobiology, School of Physics and Astronomy, University of Edinburgh, Edinburgh EH9 3FD, UK
- Division of Microbial Ecology, Department of Microbiology and Ecosystem Science, Research Network 'Chemistry Meets Microbiology', University of Vienna, Vienna 1090, Austria
| | - Roey Angel
- Division of Microbial Ecology, Department of Microbiology and Ecosystem Science, Research Network 'Chemistry Meets Microbiology', University of Vienna, Vienna 1090, Austria
| | - Charles S Cockell
- UK Centre for Astrobiology, School of Physics and Astronomy, University of Edinburgh, Edinburgh EH9 3FD, UK
| |
Collapse
|
145
|
Das S, Baker AB. Biomaterials and Nanotherapeutics for Enhancing Skin Wound Healing. Front Bioeng Biotechnol 2016; 4:82. [PMID: 27843895 PMCID: PMC5087310 DOI: 10.3389/fbioe.2016.00082] [Citation(s) in RCA: 176] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 10/11/2016] [Indexed: 02/06/2023] Open
Abstract
Wound healing is an intricate process that requires complex coordination between many cell types and an appropriate extracellular microenvironment. Chronic wounds often suffer from high protease activity, persistent infection, excess inflammation, and hypoxia. While there has been intense investigation to find new methods to improve cutaneous wound care, the management of chronic wounds, burns, and skin wound infection remain challenging clinical problems. Ideally, advanced wound dressings can provide enhanced healing and bridge the gaps in the healing processes that prevent chronic wounds from healing. These technologies have great potential for improving outcomes in patients with poorly healing wounds but face significant barriers in addressing the heterogeneity and clinical complexity of chronic or severe wounds. Active wound dressings aim to enhance the natural healing process and work to counter many aspects that plague poorly healing wounds, including excessive inflammation, ischemia, scarring, and wound infection. This review paper discusses recent advances in the development of biomaterials and nanoparticle therapeutics to enhance wound healing. In particular, this review focuses on the novel cutaneous wound treatments that have undergone significant preclinical development or are currently used in clinical practice.
Collapse
Affiliation(s)
- Subhamoy Das
- Department of Biomedical Engineering, University of Texas at Austin , Austin, TX , USA
| | - Aaron B Baker
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA; Institute for Biomaterials, Drug Delivery and Regenerative Medicine, University of Texas at Austin, Austin, TX, USA; Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, USA; Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
146
|
Kim SH, Cavaleiro AM, Rennig M, Nørholm MHH. SEVA Linkers: A Versatile and Automatable DNA Backbone Exchange Standard for Synthetic Biology. ACS Synth Biol 2016; 5:1177-1181. [PMID: 26917044 DOI: 10.1021/acssynbio.5b00257] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
DNA vectors serve to maintain and select recombinant DNA in cell factories, and as design complexity increases, there is a greater need for well-characterized parts and methods for their assembly. Standards in synthetic biology are top priority, but standardizing molecular cloning contrasts flexibility, and different researchers prefer and master different molecular technologies. Here, we describe a new, highly versatile and automatable standard "SEVA linkers" for vector exchange. SEVA linkers enable backbone swapping with 20 combinations of classical enzymatic restriction/ligation, Gibson isothermal assembly, uracil excision cloning, and a nicking enzyme-based methodology we term SEVA cloning. SEVA cloning is a simplistic one-tube protocol for backbone swapping directly from plasmid stock solutions. We demonstrate the different performance of 30 plasmid backbones for small molecule and protein production and obtain more than 10-fold improvement from a four-gene biosynthetic pathway and 430-fold improvement with a difficult-to-express membrane protein. The standardized linkers and protocols add to the Standard European Vectors Architecture (SEVA) resource and are freely available to the synthetic biology community.
Collapse
Affiliation(s)
- Se Hyeuk Kim
- Novo Nordisk Foundation Center
for Biosustainability, Technical University of Denmark, Hørsholm DK-2970, Denmark
| | - Ana Mafalda Cavaleiro
- Novo Nordisk Foundation Center
for Biosustainability, Technical University of Denmark, Hørsholm DK-2970, Denmark
| | - Maja Rennig
- Novo Nordisk Foundation Center
for Biosustainability, Technical University of Denmark, Hørsholm DK-2970, Denmark
| | - Morten H. H. Nørholm
- Novo Nordisk Foundation Center
for Biosustainability, Technical University of Denmark, Hørsholm DK-2970, Denmark
| |
Collapse
|
147
|
Khan MA, Göpel Y, Milewski S, Görke B. Two Small RNAs Conserved in Enterobacteriaceae Provide Intrinsic Resistance to Antibiotics Targeting the Cell Wall Biosynthesis Enzyme Glucosamine-6-Phosphate Synthase. Front Microbiol 2016; 7:908. [PMID: 27379045 PMCID: PMC4908143 DOI: 10.3389/fmicb.2016.00908] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 05/27/2016] [Indexed: 11/17/2022] Open
Abstract
Formation of glucosamine-6-phosphate (GlcN6P) by enzyme GlcN6P synthase (GlmS) represents the first step in bacterial cell envelope synthesis. In Escherichia coli, expression of glmS is controlled by small RNAs (sRNAs) GlmY and GlmZ. GlmZ activates the glmS mRNA by base-pairing. When not required, GlmZ is bound by adapter protein RapZ and recruited to cleavage by RNase E inactivating the sRNA. The homologous sRNA GlmY activates glmS indirectly. When present at high levels, GlmY sequesters RapZ by an RNA mimicry mechanism suppressing cleavage of GlmZ. The interplay of both sRNAs is believed to adjust GlmS synthesis to the needs of the cell, i.e., to achieve GlcN6P homeostasis. Bacilysin (tetaine) and Nva-FMDP are dipeptide antibiotics that impair cell envelope synthesis by inhibition of enzyme GlmS through covalent modification. However, although taken up efficiently, these antibiotics are less active against E. coli for reasons unknown so far. Here we show that the GlmY/GlmZ circuit provides resistance. Inhibition of GlmS causes GlcN6P deprivation leading to activation of GlmY and GlmZ, which in turn trigger glmS overexpression in a dosage-dependent manner. Mutation of glmY or glmZ disables this response and renders the bacteria highly susceptible to GlmS inhibitors. Thus, E. coli compensates inhibition of GlmS by increasing its synthesis through the GlmY/GlmZ pathway. This mechanism is also operative in Salmonella indicating that it is conserved in Enterobacteriaceae possessing these sRNAs. As GlmY apparently responds to GlcN6P, co-application of a non-metabolizable GlcN6P analog may prevent activation of the sRNAs and thereby increase the bactericidal activity of GlmS inhibitors against wild-type bacteria. Initial experiments using glucosamine-6-sulfate support this possibility. Thus, GlcN6P analogs might be considered for co-application with GlmS inhibitors in combined therapy to treat infections caused by pathogenic Enterobacteriaceae.
Collapse
Affiliation(s)
- Muna A. Khan
- Department of Microbiology, Immunobiology and Genetics, Max F. Perutz Laboratories, University of Vienna, Vienna BiocenterVienna, Austria
| | - Yvonne Göpel
- Department of Microbiology, Immunobiology and Genetics, Max F. Perutz Laboratories, University of Vienna, Vienna BiocenterVienna, Austria
| | - Slawomir Milewski
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdańsk University of TechnologyGdańsk, Poland
| | - Boris Görke
- Department of Microbiology, Immunobiology and Genetics, Max F. Perutz Laboratories, University of Vienna, Vienna BiocenterVienna, Austria
| |
Collapse
|
148
|
The analysis of the antibiotic resistome offers new opportunities for therapeutic intervention. Future Med Chem 2016; 8:1133-51. [DOI: 10.4155/fmc-2016-0027] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Most efforts in the development of antimicrobials have focused on the screening of lethal targets. Nevertheless, the constant expansion of antimicrobial resistance makes the antibiotic resistance determinants themselves suitable targets for finding inhibitors to be used in combination with antibiotics. Among them, inhibitors of antibiotic inactivating enzymes and of multidrug efflux pumps are suitable candidates for improving the efficacy of antibiotics. In addition, the application of systems biology tools is helping to understand the changes in bacterial physiology associated to the acquisition of resistance, including the increased susceptibility to other antibiotics displayed by some antibiotic-resistant mutants. This information is useful for implementing novel strategies based in metabolic interventions or combination of antibiotics for improving the efficacy of antibacterial therapy.
Collapse
|
149
|
Larrouy-Maumus G, Marino LB, Madduri AVR, Ragan TJ, Hunt DM, Bassano L, Gutierrez MG, Moody DB, Pavan FR, de Carvalho LPS. Cell-Envelope Remodeling as a Determinant of Phenotypic Antibacterial Tolerance in Mycobacterium tuberculosis. ACS Infect Dis 2016; 2:352-360. [PMID: 27231718 PMCID: PMC4877114 DOI: 10.1021/acsinfecdis.5b00148] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Indexed: 12/03/2022]
Abstract
![]()
The mechanisms that lead to phenotypic
antibacterial tolerance in bacteria remain poorly understood. We investigate
whether changes in NaCl concentration toward physiologically higher
values affect antibacterial efficacy against Mycobacterium
tuberculosis (Mtb), the causal agent of human tuberculosis.
Indeed, multiclass phenotypic antibacterial tolerance is observed
during Mtb growth in physiologic saline. This includes changes in
sensitivity to ethionamide, ethambutol, d-cycloserine, several
aminoglycosides, and quinolones. By employing organism-wide metabolomic
and lipidomic approaches combined with phenotypic tests, we identified
a time-dependent biphasic adaptive response after exposure of Mtb
to physiological levels of NaCl. A first rapid, extensive, and reversible
phase was associated with changes in core and amino acid metabolism.
In a second phase, Mtb responded with a substantial remodelling of
plasma membrane and outer lipid membrane composition. We demonstrate
that phenotypic tolerance at physiological concentrations of NaCl
is the result of changes in plasma and outer membrane lipid remodeling
and not changes in core metabolism. Altogether, these results indicate
that physiologic saline-induced antibacterial tolerance is kinetically
coupled to cell envelope changes and demonstrate that metabolic changes
and growth arrest are not the cause of phenotypic tolerance observed
in Mtb exposed to physiologic concentrations of NaCl. Importantly,
this work uncovers a role for bacterial cell envelope remodeling in
antibacterial tolerance, alongside well-documented allterations in
respiration, metabolism, and growth rate.
Collapse
Affiliation(s)
- Gérald Larrouy-Maumus
- Laboratory
of Chemical Biology of Tuberculosis Pathogenesis, MRC Centre for Molecular
Bacteriology and Infection, Imperial College London, Kensington, London SW7 2DD, United Kingdom
| | - Leonardo B. Marino
- School of Pharmaceutical
Sciences, São Paulo State University (UNESP), 4801-902 Araraquara, SP, Brazil
| | - Ashoka V. R. Madduri
- Division of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts 02115, United States
| | | | | | - Lucrezia Bassano
- Laboratory
of Chemical Biology of Tuberculosis Pathogenesis, MRC Centre for Molecular
Bacteriology and Infection, Imperial College London, Kensington, London SW7 2DD, United Kingdom
| | | | - D. Branch Moody
- Division of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Fernando R. Pavan
- School of Pharmaceutical
Sciences, São Paulo State University (UNESP), 4801-902 Araraquara, SP, Brazil
| | | |
Collapse
|
150
|
OsdR of Streptomyces coelicolor and the Dormancy Regulator DevR of Mycobacterium tuberculosis Control Overlapping Regulons. mSystems 2016; 1:mSystems00014-16. [PMID: 27822533 PMCID: PMC5069765 DOI: 10.1128/msystems.00014-16] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 03/29/2016] [Indexed: 11/20/2022] Open
Abstract
Dormancy is a state of growth cessation that allows bacteria to escape the host defense system and antibiotic challenge. Understanding the mechanisms that control dormancy is of key importance for the treatment of latent infections, such as those from Mycobacterium tuberculosis. In mycobacteria, dormancy is controlled by the response regulator DevR, which responds to conditions of hypoxia. Here, we show that OsdR of Streptomyces coelicolor recognizes the same regulatory element and controls a regulon that consists of genes involved in the control of stress and development. Only the core regulon in the direct vicinity of dosR and osdR is conserved between M. tuberculosis and S. coelicolor, respectively. Thus, we show how the system has diverged from allowing escape from the host defense system by mycobacteria to the control of sporulation by complex multicellular streptomycetes. This provides novel insights into how bacterial growth and development are coordinated with the environmental conditions. Two-component regulatory systems allow bacteria to respond adequately to changes in their environment. In response to a given stimulus, a sensory kinase activates its cognate response regulator via reversible phosphorylation. The response regulator DevR activates a state of dormancy under hypoxia in Mycobacterium tuberculosis, allowing this pathogen to escape the host defense system. Here, we show that OsdR (SCO0204) of the soil bacterium Streptomyces coelicolor is a functional orthologue of DevR. OsdR, when activated by the sensory kinase OsdK (SCO0203), binds upstream of the DevR-controlled dormancy genes devR, hspX, and Rv3134c of M. tuberculosis. In silico analysis of the S. coelicolor genome combined with in vitro DNA binding studies identified many binding sites in the genomic region around osdR itself and upstream of stress-related genes. This binding correlated well with transcriptomic responses, with deregulation of developmental genes and genes related to stress and hypoxia in the osdR mutant. A peak in osdR transcription in the wild-type strain at the onset of aerial growth correlated with major changes in global gene expression. Taken together, our data reveal the existence of a dormancy-related regulon in streptomycetes which plays an important role in the transcriptional control of stress- and development-related genes. IMPORTANCE Dormancy is a state of growth cessation that allows bacteria to escape the host defense system and antibiotic challenge. Understanding the mechanisms that control dormancy is of key importance for the treatment of latent infections, such as those from Mycobacterium tuberculosis. In mycobacteria, dormancy is controlled by the response regulator DevR, which responds to conditions of hypoxia. Here, we show that OsdR of Streptomyces coelicolor recognizes the same regulatory element and controls a regulon that consists of genes involved in the control of stress and development. Only the core regulon in the direct vicinity of dosR and osdR is conserved between M. tuberculosis and S. coelicolor, respectively. Thus, we show how the system has diverged from allowing escape from the host defense system by mycobacteria to the control of sporulation by complex multicellular streptomycetes. This provides novel insights into how bacterial growth and development are coordinated with the environmental conditions.
Collapse
|